Network


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

Hotspot


Dive into the research topics where Steven Grinnell is active.

Publication


Featured researches published by Steven Grinnell.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Truncated G protein-coupled mu opioid receptor MOR-1 splice variants are targets for highly potent opioid analgesics lacking side effects

Susruta Majumdar; Steven Grinnell; Valerie Le Rouzic; Maxim Burgman; Lisa Polikar; Michael Ansonoff; John E. Pintar; Ying-Xian Pan; Gavril W. Pasternak

Pain remains a pervasive problem throughout medicine, transcending all specialty boundaries. Despite the extraordinary insights into pain and its mechanisms over the past few decades, few advances have been made with analgesics. Most pain remains treated by opiates, which have significant side effects that limit their utility. We now describe a potent opiate analgesic lacking the traditional side effects associated with classical opiates, including respiratory depression, significant constipation, physical dependence, and, perhaps most important, reinforcing behavior, demonstrating that it is possible to dissociate side effects from analgesia. Evidence indicates that this agent acts through a truncated, six-transmembrane variant of the G protein-coupled mu opioid receptor MOR-1. Although truncated splice variants have been reported for a number of G protein-coupled receptors, their functional relevance has been unclear. Our evidence now suggests that truncated variants can be physiologically important through heterodimerization, even when inactive alone, and can comprise new therapeutic targets, as illustrated by our unique opioid analgesics with a vastly improved pharmacological profile.


Bioorganic & Medicinal Chemistry Letters | 2011

Generation of novel radiolabeled opiates through site-selective iodination.

Susruta Majumdar; Maxim Burgman; Nathan Haselton; Steven Grinnell; Julia Ocampo; Anna R. Pasternak; Gavril W. Pasternak

Tritiated opioid radioligands have proven valuable in exploring opioid binding sites. However, tritium has many limitations. Its low specific activity and limited counting efficiency makes it difficult to examine low abundant, high affinity sites and its disposal is problematic due to the need to use organic scintillants and its relatively long half-life. To overcome these issues, we have synthesized both unlabeled and carrier-free radioiodinated iodobenzoyl derivatives of 6β-naltrexamine ((125)I-BNtxA, 18), 6β-naloxamine ((125)I-BNalA, 19) and 6β-oxymorphamine ((125)I-BOxyA, 20) with specific activities of 2100Ci/mmol. To optimize the utility of the radioligand, we designed a synthesis in which the radiolabel is incorporated in the last synthetic step, which required the selective iodination of the benzoyl moiety without incorporation into the phenolic A ring. Competition studies demonstrated high affinity of the unlabelled compounds for opioid receptors in transfected cell lines, as did the direct binding of the (125)I-ligands to the opioid receptors. The radioligand displayed very high sensitivity, enabling a marked reduction in tissue, as well as excellent signal/noise characteristics. These new (125)I-radioligands should prove valuable in future studies of opioid binding sites.


Neuropsychopharmacology | 2017

The Behavioral Effects of the Antidepressant Tianeptine Require the Mu-Opioid Receptor

Benjamin Adam Samuels; Katherine M. Nautiyal; Andrew C. Kruegel; Marjorie R Levinstein; Valerie M Magalong; Madalee M. Gassaway; Steven Grinnell; Jaena Han; Michael Ansonoff; John E. Pintar; Jonathan A. Javitch; Dalibor Sames; René Hen

Depression is a debilitating chronic illness that affects around 350 million people worldwide. Current treatments, such as selective serotonin reuptake inhibitors, are not ideal because only a fraction of patients achieve remission. Tianeptine is an effective antidepressant with a previously unknown mechanism of action. We recently reported that tianeptine is a full agonist at the mu opioid receptor (MOR). Here we demonstrate that the acute and chronic antidepressant-like behavioral effects of tianeptine in mice require MOR. Interestingly, while tianeptine also produces many opiate-like behavioral effects such as analgesia and reward, it does not lead to tolerance or withdrawal. Furthermore, the primary metabolite of tianeptine (MC5), which has a longer half-life, mimics the behavioral effects of tianeptine in a MOR-dependent fashion. These results point to the possibility that MOR and its downstream signaling cascades may be novel targets for antidepressant drug development.


Journal of Pharmacology and Experimental Therapeutics | 2014

Pharmacologic Characterization in the Rat of a Potent Analgesic Lacking Respiratory Depression, IBNtxA

Steven Grinnell; Susruta Majumdar; Ankita Narayan; V. Le Rouzic; Michael Ansonoff; John E. Pintar; Gavril W. Pasternak

IBNtxA (3′-iodobenzoyl-6β-naltrexamide) is a potent analgesic in mice lacking many traditional opioid side effects. In mice, it displays no respiratory depression, does not produce physical dependence with chronic administration, and shows no cross-tolerance to morphine. It has limited effects on gastrointestinal transit and shows no reward behavior. Biochemical studies indicate its actions are mediated through a set of μ-opioid receptor clone MOR-1 splice variants associated with exon 11 that lack exon 1 and contain only six transmembrane domains. Like the mouse and human, rats express exon 11–associated splice variants that also contain only six transmembrane domains, raising the question of whether IBNtxA would have a similar pharmacologic profile in rats. When given systemically, IBNtxA is a potent analgesic in rats, with an ED50 value of 0.89 mg/kg s.c., approximately 4-fold more potent than morphine. It shows no analgesic cross-tolerance in morphine-pelleted rats. IBNtxA displays no respiratory depression as measured by blood oxygen saturation. In contrast, oximetry shows that an equianalgesic dose of morphine lowers blood oxygen saturation values by 30%. IBNtxA binding is present in a number of brain regions, with the thalamus standing out with very high levels and the cerebellum with low levels. As in mice, IBNtxA is a potent analgesic in rats with a favorable pharmacologic profile and reduced side effects.


Synapse | 2016

Mediation of buprenorphine analgesia by a combination of traditional and truncated mu opioid receptor splice variants

Steven Grinnell; Michael Ansonoff; Gina F. Marrone; Zhigang Lu; Ankita Narayan; Jin Xu; Grace C. Rossi; Susruta Majumdar; Ying-Xian Pan; Daniel L. Bassoni; John E. Pintar; Gavril W. Pasternak

Buprenorphine has long been classified as a mu analgesic, although its high affinity for other opioid receptor classes and the orphanin FQ/nociceptin ORL1 receptor may contribute to its other actions. The current studies confirmed a mu mechanism for buprenorphine analgesia, implicating several subsets of mu receptor splice variants. Buprenorphine analgesia depended on the expression of both exon 1‐associated traditional full length 7 transmembrane (7TM) and exon 11‐associated truncated 6 transmembrane (6TM) MOR‐1 variants. In genetic models, disruption of delta, kappa1 or ORL1 receptors had no impact on buprenorphine analgesia, while loss of the traditional 7TM MOR‐1 variants in an exon 1 knockout (KO) mouse markedly lowered buprenorphine analgesia. Loss of the truncated 6TM variants in an exon 11 KO mouse totally eliminated buprenorphine analgesia. In distinction to analgesia, the inhibition of gastrointestinal transit and stimulation of locomotor activity were independent of truncated 6TM variants. Restoring expression of a 6TM variant with a lentivirus rescued buprenorphine analgesia in an exon 11 KO mouse that still expressed the 7TM variants. Despite a potent and robust stimulation of 35S‐GTPγS binding in MOR‐1 expressing CHO cells, buprenorphine failed to recruit β‐arrestin‐2 binding at doses as high as 10 µM. Buprenorphine was an antagonist in DOR‐1 expressing cells and an inverse agonist in KOR‐1 cells. Buprenorphine analgesia is complex and requires multiple mu receptor splice variant classes but other actions may involve alternative receptors.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Truncated mu opioid GPCR variant involvement in opioid-dependent and opioid-independent pain modulatory systems within the CNS

Gina F. Marrone; Steven Grinnell; Zhigang Lu; Grace C. Rossi; Valerie Le Rouzic; Jin Xu; Susruta Majumdar; Ying-Xian Pan; Gavril W. Pasternak

Significance Many classes of G protein-coupled receptors (GPCRs) produce truncated variants. Truncated forms of the mu opioid receptor gene Oprm1 containing only six transmembrane domains (6TM) can mediate a potent analgesia without producing many classical opioid side effects. We now show that 6TM Oprm1 splice variants are essential in the analgesic actions of delta and kappa opioids as well as α2 adrenergic drugs, but not neurotensin, cannabinoids, or muscarinic drugs. The role of the 6TM variants seems limited to analgesia because they are not involved with kappa aversion, delta-induced seizure activity, or α2 adrenergic hypolocomotion. These findings emphasize the importance of 6TM Oprm1 variants in opioid and nonopioid sensory processing and illustrate the potential importance of the vast array of other classes of truncated GPCR variants. The clinical management of severe pain depends heavily on opioids acting through mu opioid receptors encoded by the Oprm1 gene, which undergoes extensive alternative splicing. In addition to generating a series of prototypic seven transmembrane domain (7TM) G protein-coupled receptors (GPCRs), Oprm1 also produces a set of truncated splice variants containing only six transmembrane domains (6TM) through which selected opioids such as IBNtxA (3′-iodobenzoyl-6β-naltrexamide) mediate a potent analgesia without many undesirable effects. Although morphine analgesia is independent of these 6TM mu receptor isoforms, we now show that the selective loss of the 6TM variants in a knockout model eliminates the analgesic actions of delta and kappa opioids and of α2-adrenergic compounds, but not cannabinoid, neurotensin, or muscarinic drugs. These observations were confirmed by using antisense paradigms. Despite their role in analgesia, loss of the 6TM variants were not involved with delta opioid-induced seizure activity, aversion to the kappa drug U50,488H, or α2-mediated hypolocomotion. These observations support the existence of parallel opioid and nonopioid pain modulatory systems and highlight the ability to dissociate unwanted delta, kappa1, and α2 actions from analgesia.


Bioorganic & Medicinal Chemistry Letters | 2013

Sandmeyer reaction repurposed for the site-selective, non-oxidizing radioiodination of fully-deprotected peptides: Studies on the endogenous opioid peptide α-neoendorphin

Julie E. Pickett; Kunihiko Nagakura; Anna R. Pasternak; Steven Grinnell; Susruta Majumdar; Jason S. Lewis; Gavril W. Pasternak

Standard radioiodination methods lack site-selectivity and either mask charges (Bolton-Hunter) or involve oxidative reaction conditions (chloramine-T). Opioid peptides are very sensitive to certain structural modifications, making these labeling methods untenable. In our model opioid peptide, α-neoendorphin, we replaced a tyrosyl hydroxyl with an iodine, and in cell lines stably expressing mu, delta, or kappa opioid receptors, we saw no negative effects on binding. We then optimized a repurposed Sandmeyer reaction using copper(I) catalysts with non-redoxing/non-nucleophilic ligands, bringing the radiochemical yield up to around 30%, and site-selectively incorporated radioactive iodine into this position under non-oxidizing reaction conditions, which should be broadly compatible with most peptides. The (125)I- and (131)I-labeled versions of the compound bound with high affinity to opioid receptors in mouse brain homogenates, thus demonstrating the general utility of the labeling strategy and of the peptide for exploring opioid binding sites.


Journal of Medicinal Chemistry | 2012

Synthesis and Evaluation of Aryl-Naloxamide Opiate Analgesics Targeting Truncated Exon 11-Associated μ Opioid Receptor (MOR-1) Splice Variants

Susruta Majumdar; Joan J. Subrath; Valerie Le Rouzic; Lisa Polikar; Maxim Burgman; Kuni Nagakura; Julie Ocampo; Nathan Haselton; Anna R. Pasternak; Steven Grinnell; Ying-Xian Pan; Gavril W. Pasternak


The FASEB Journal | 2015

A novel opioid for photoaffinity labeling opioid receptor complexes in natively-expressing tissues

Steven Grinnell; Susruta Majumdar; Ankita Narayan; Gavril W. Pasternak


The FASEB Journal | 2015

Endomorphins and DAMGO Utilize Different Mechanisms to Produce Analgesia

Gina F. Marrone; Steven Grinnell; Grace C. Rossi; John E. Pintar; Susruta Majumdar; Ying-Xian Pan; Gavril W. Pasternak

Collaboration


Dive into the Steven Grinnell's collaboration.

Top Co-Authors

Avatar

Gavril W. Pasternak

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Susruta Majumdar

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ying-Xian Pan

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

John E. Pintar

University of Medicine and Dentistry of New Jersey

View shared research outputs
Top Co-Authors

Avatar

Michael Ansonoff

University of Medicine and Dentistry of New Jersey

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna R. Pasternak

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maxim Burgman

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge