1
|
Asp AJ, Boschen SL, Chang SY, Kim J, Silvernail JL, Lujan JL. An ultra low frequency spike timing dependent plasticity based approach for reducing alcohol drinking. Sci Rep 2024; 14:30907. [PMID: 39730615 DOI: 10.1038/s41598-024-81390-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/26/2024] [Indexed: 12/29/2024] Open
Abstract
Alcohol use disorder (AUD) is a chronic relapsing brain disorder characterized by an impaired ability to stop or control alcohol consumption despite adverse social, occupational, or health consequences. AUD affects nearly one-third of adults at some point during their lives, with an associated cost of approximately $249 billion annually in the U.S. alone. The effects of alcohol consumption are expected to increase significantly during the COVID-19 pandemic, with alcohol sales increasing by approximately 54%, potentially exacerbating health concerns and risk-taking behaviors. Unfortunately, existing pharmacological and behavioral therapies for AUD are associated with poor success rates, with approximately 40% of individuals relapsing within three years of treatment.Pre-clinical studies have shown that chronic alcohol consumption leads to significant changes in synaptic function within the dorsal medial striatum (DMS), one of the brain regions associated with AUD and responsible for mediating goal-directed behavior. Specifically, chronic alcohol consumption has been associated with hyperactivity of dopamine receptor 1 (D1) medium spiny neurons (MSN) and hypoactivity of dopamine receptor 2 (D1) MSNs within the DMS. Optogenetic, chemogenetic, and transgenic approaches have demonstrated that reducing the D1/D2 MSN signaling imbalance decreases alcohol self-administration in rodent models of AUD.Here, we present an electrical stimulation approach that uses ultra-low (≤ 1 Hz) frequency (ULF) spike-timing-dependent plasticity (STDP) in mouse models of AUD to reduce DMS D1/D2 MSN signaling imbalances by stimulating D1-MSN afferents into the GPi and ACC glutamatergic projections to the DMS in a time-locked stimulation sequence. Our data suggest that GPi/ACC ULF-STDP selectively decreases DMS D1-MSN hyperactivity leading to reduced alcohol consumption without evoking undesired affective behaviors using electrical stimulation rather than approaches requiring genetic modification. This work represents a step towards fulfilling the unmet need for a reliable method of treating severe AUD through cell-type-specific control with clinically available neuromodulation tools.
Collapse
Affiliation(s)
- Anders J Asp
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, 55905, USA
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Su-Youne Chang
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jiwon Kim
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jodi L Silvernail
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - J Luis Lujan
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
2
|
Neel AI, Wang Y, Sun H, Liontis KE, McCormack MC, Mayer JC, Cervera Juanes RP, Davenport AT, Grant KA, Daunais JD, Chen R. Differential regulation of G protein-coupled receptor-associated proteins in the caudate and the putamen of cynomolgus macaques following chronic ethanol drinking. J Neurochem 2024; 168:2722-2735. [PMID: 38783749 PMCID: PMC11449652 DOI: 10.1111/jnc.16134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/16/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
The dorsal striatum is composed of the caudate nucleus and the putamen in human and non-human primates. These two regions receive different cortical projections and are functionally distinct. The caudate is involved in the control of goal-directed behaviors, while the putamen is implicated in habit learning and formation. Previous reports indicate that ethanol differentially influences neurotransmission in these two regions. Because neurotransmitters primarily signal through G protein-coupled receptors (GPCRs) to modulate neuronal activity, the present study aimed to determine whether ethanol had a region-dependent impact on the expression of proteins that are involved in the trafficking and function of GPCRs, including G protein subunits and their effectors, protein kinases, and elements of the cytoskeleton. Western blotting was performed to examine protein levels in the caudate and the putamen of male cynomolgus macaques that self-administered ethanol for 1 year under free access conditions, along with control animals that self-administered an isocaloric sweetened solution under identical operant conditions. Among the 18 proteins studied, we found that the levels of one protein (PKCβ) were increased, and 13 proteins (Gαi1/3, Gαi2, Gαo, Gβ1γ, PKCα, PKCε, CaMKII, GSK3β, β-actin, cofilin, α-tubulin, and tubulin polymerization promoting protein) were reduced in the caudate of alcohol-drinking macaques. However, ethanol did not alter the expression of any proteins examined in the putamen. These observations underscore the unique vulnerability of the caudate nucleus to changes in protein expression induced by chronic ethanol exposure. Whether these alterations are associated with ethanol-induced dysregulation of GPCR function and neurotransmission warrants future investigation.
Collapse
Affiliation(s)
- Anna I. Neel
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| | - Yutong Wang
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| | - Haiguo Sun
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| | - Katherine E. Liontis
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| | - Mary C. McCormack
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| | - Jonathan C. Mayer
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| | - Rita P. Cervera Juanes
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| | - April T. Davenport
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| | - Kathleen A. Grant
- Division of Neuroscience Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR 97239
| | - James D. Daunais
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| | - Rong Chen
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston Salem, NC 27157
| |
Collapse
|
3
|
Meqbil YJ, Aguilar J, Blaine AT, Chen L, Cassell RJ, Pradhan AA, van Rijn RM. Identification of 1,3,8-Triazaspiro[4.5]Decane-2,4-Dione Derivatives as a Novel δ Opioid Receptor-Selective Agonist Chemotype. J Pharmacol Exp Ther 2024; 389:301-309. [PMID: 38621994 PMCID: PMC11125782 DOI: 10.1124/jpet.123.001735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 03/25/2024] [Accepted: 04/08/2024] [Indexed: 04/17/2024] Open
Abstract
δ opioid receptors (DORs) hold potential as a target for neurologic and psychiatric disorders, yet no DOR agonist has proven efficacious in critical phase II clinical trials. The exact reasons for the failure to produce quality drug candidates for the DOR are unclear. However, it is known that certain DOR agonists can induce seizures and exhibit tachyphylaxis. Several studies have suggested that those adverse effects are more prevalent in delta agonists that share the (+)-4-[(αR)-α-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide (SNC80)/4-[(αR*)-α-((2S*,5R*)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-hydroxybenzyl]-N,N-diethylbenzamide chemotype. There is a need to find novel lead candidates for drug development that have improved pharmacological properties to differentiate them from the current failed delta agonists. Our objective in this study was to identify novel DOR agonists. We used a β-arrestin assay to screen a small G-protein coupled receptors (GPCR)-focused chemical library. We identified a novel chemotype of DOR agonists that appears to bind to the orthosteric site based of docking and molecular dynamic simulation. The most potent agonist hit compound is selective for the DOR over a panel of 167 other GPCRs, is slightly biased toward G-protein signaling and has anti-allodynic efficacy in a complete Freund's adjuvant model of inflammatory pain in C57BL/6 male and female mice. The newly discovered chemotype contrasts with molecules like SNC80 that are highly efficacious β-arrestin recruiters and may suggest this novel class of DOR agonists could be expanded on to develop a clinical candidate drug. SIGNIFICANCE STATEMENT: δ opioid receptors are a clinical target for various neurological disorders, including migraine and chronic pain. Many of the clinically tested delta opioid agonists share a single chemotype, which carries risks during drug development. Through a small-scale high-throughput screening assay, this study identified a novel δ opioid receptor agonist chemotype, which may serve as alternative for the current analgesic clinical candidates.
Collapse
Affiliation(s)
- Yazan J Meqbil
- Borch Department of Medicinal Chemistry and Molecular Pharmacology (Y.J.M., A.T.B., R.J.C., R.M.v.R.), Computational Interdisciplinary Graduate Programs, Computational Life Sciences (Y.J.M.), and Interdisciplinary Life Science-PULSe (A.T.B.), Purdue University, West Lafayette, Indiana; Purdue Institute for Integrative Neuroscience, West Lafayette, Indiana (R.M.v.R.); Purdue Institute for Drug Discovery, West Lafayette, Indiana (L.C., R.M.v.R.); Septerna Inc., South San Francisco, California (R.M.v.R.); and Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri (J.A., A.A.P.)
| | - Jhoan Aguilar
- Borch Department of Medicinal Chemistry and Molecular Pharmacology (Y.J.M., A.T.B., R.J.C., R.M.v.R.), Computational Interdisciplinary Graduate Programs, Computational Life Sciences (Y.J.M.), and Interdisciplinary Life Science-PULSe (A.T.B.), Purdue University, West Lafayette, Indiana; Purdue Institute for Integrative Neuroscience, West Lafayette, Indiana (R.M.v.R.); Purdue Institute for Drug Discovery, West Lafayette, Indiana (L.C., R.M.v.R.); Septerna Inc., South San Francisco, California (R.M.v.R.); and Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri (J.A., A.A.P.)
| | - Arryn T Blaine
- Borch Department of Medicinal Chemistry and Molecular Pharmacology (Y.J.M., A.T.B., R.J.C., R.M.v.R.), Computational Interdisciplinary Graduate Programs, Computational Life Sciences (Y.J.M.), and Interdisciplinary Life Science-PULSe (A.T.B.), Purdue University, West Lafayette, Indiana; Purdue Institute for Integrative Neuroscience, West Lafayette, Indiana (R.M.v.R.); Purdue Institute for Drug Discovery, West Lafayette, Indiana (L.C., R.M.v.R.); Septerna Inc., South San Francisco, California (R.M.v.R.); and Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri (J.A., A.A.P.)
| | - Lan Chen
- Borch Department of Medicinal Chemistry and Molecular Pharmacology (Y.J.M., A.T.B., R.J.C., R.M.v.R.), Computational Interdisciplinary Graduate Programs, Computational Life Sciences (Y.J.M.), and Interdisciplinary Life Science-PULSe (A.T.B.), Purdue University, West Lafayette, Indiana; Purdue Institute for Integrative Neuroscience, West Lafayette, Indiana (R.M.v.R.); Purdue Institute for Drug Discovery, West Lafayette, Indiana (L.C., R.M.v.R.); Septerna Inc., South San Francisco, California (R.M.v.R.); and Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri (J.A., A.A.P.)
| | - Robert J Cassell
- Borch Department of Medicinal Chemistry and Molecular Pharmacology (Y.J.M., A.T.B., R.J.C., R.M.v.R.), Computational Interdisciplinary Graduate Programs, Computational Life Sciences (Y.J.M.), and Interdisciplinary Life Science-PULSe (A.T.B.), Purdue University, West Lafayette, Indiana; Purdue Institute for Integrative Neuroscience, West Lafayette, Indiana (R.M.v.R.); Purdue Institute for Drug Discovery, West Lafayette, Indiana (L.C., R.M.v.R.); Septerna Inc., South San Francisco, California (R.M.v.R.); and Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri (J.A., A.A.P.)
| | - Amynah A Pradhan
- Borch Department of Medicinal Chemistry and Molecular Pharmacology (Y.J.M., A.T.B., R.J.C., R.M.v.R.), Computational Interdisciplinary Graduate Programs, Computational Life Sciences (Y.J.M.), and Interdisciplinary Life Science-PULSe (A.T.B.), Purdue University, West Lafayette, Indiana; Purdue Institute for Integrative Neuroscience, West Lafayette, Indiana (R.M.v.R.); Purdue Institute for Drug Discovery, West Lafayette, Indiana (L.C., R.M.v.R.); Septerna Inc., South San Francisco, California (R.M.v.R.); and Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri (J.A., A.A.P.)
| | - Richard M van Rijn
- Borch Department of Medicinal Chemistry and Molecular Pharmacology (Y.J.M., A.T.B., R.J.C., R.M.v.R.), Computational Interdisciplinary Graduate Programs, Computational Life Sciences (Y.J.M.), and Interdisciplinary Life Science-PULSe (A.T.B.), Purdue University, West Lafayette, Indiana; Purdue Institute for Integrative Neuroscience, West Lafayette, Indiana (R.M.v.R.); Purdue Institute for Drug Discovery, West Lafayette, Indiana (L.C., R.M.v.R.); Septerna Inc., South San Francisco, California (R.M.v.R.); and Center for Clinical Pharmacology, Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri (J.A., A.A.P.)
| |
Collapse
|
4
|
Kajino K, Tokuda A, Saitoh T. Morphinan Evolution: The Impact of Advances in Biochemistry and Molecular Biology. J Biochem 2024; 175:337-355. [PMID: 38382631 DOI: 10.1093/jb/mvae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/01/2024] [Accepted: 02/09/2024] [Indexed: 02/23/2024] Open
Abstract
Morphinan-based opioids, derived from natural alkaloids like morphine, codeine and thebaine, have long been pivotal in managing severe pain. However, their clinical utility is marred by significant side effects and high addiction potential. This review traces the evolution of the morphinan scaffold in light of advancements in biochemistry and molecular biology, which have expanded our understanding of opioid receptor pharmacology. We explore the development of semi-synthetic and synthetic morphinans, their receptor selectivity and the emergence of biased agonism as a strategy to dissociate analgesic properties from undesirable effects. By examining the molecular intricacies of opioid receptors and their signaling pathways, we highlight how receptor-type selectivity and signaling bias have informed the design of novel analgesics. This synthesis of historical and contemporary perspectives provides an overview of the morphinan landscape, underscoring the ongoing efforts to mitigate the problems facing opioids through smarter drug design. We also highlight that most morphinan derivatives show a preference for the G protein pathway, although detailed experimental comparisons are still necessary. This fact underscores the utility of the morphinan skeleton in future opioid drug discovery.
Collapse
Affiliation(s)
- Keita Kajino
- International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Degree Programs in Pure and Applied Sciences, Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
| | - Akihisa Tokuda
- International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Doctoral Program in Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Tsuyoshi Saitoh
- International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Doctoral Program in Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
5
|
Doyle MA, Taylor A, Winder DG. Neural Circuitries and Alcohol Use Disorder: Cutting Corners in the Cycle. Curr Top Behav Neurosci 2023. [PMID: 38082108 DOI: 10.1007/7854_2023_454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2024]
Abstract
An implicit tenet of the alcohol use disorder (AUD) research field is that knowledge of how alcohol interacts with the brain is critical to the development of an understanding of vulnerability to AUD and treatment approaches. Gaining this understanding requires the mapping of brain function critical to specific components of this heterogeneous disorder. Early approaches in humans and animal models focused on the determination of specific brain regions sensitive to alcohol action and their participation in AUD-relevant behaviors. Broadly speaking, this research has focused on three domains, Binge/Intoxication, Negative Affect/Withdrawal, and Preoccupation/Anticipation, with a number of regions identified as participating in each. With the generational advances in technologies that the field of neuroscience has undergone over the last two decades, this focus has shifted to a circuit-based analysis. A wealth of new data has sharpened the field's focus on the specific roles of the interconnectivity of multiple brain regions in AUD and AUD-relevant behaviors, as well as demonstrating that the three major domains described above have much fuzzier edges than originally thought.In this chapter, we very briefly review brain regions previously implicated in aspects of AUD-relevant behavior from animal model research. Next, we move to a more in-depth overview of circuit-based approaches, and the utilization of these approaches in current AUD research.
Collapse
Affiliation(s)
- Marie A Doyle
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Anne Taylor
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Danny G Winder
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
6
|
Cruz B, Castañeda K, Aranda M, Hinojosa CA, Castro-Gutierrez R, Flores RJ, Spencer CT, Vozella V, Roberto M, Gadad BS, Roychowdhury S, O’Dell LE. Alcohol self-administration and nicotine withdrawal alter biomarkers of stress and inflammation and prefrontal cortex changes in Gβ subunits. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2023; 49:321-332. [PMID: 36206520 PMCID: PMC10348398 DOI: 10.1080/00952990.2022.2121656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/28/2022] [Accepted: 09/02/2022] [Indexed: 11/07/2022]
Abstract
Background: Although alcohol and nicotine are often used together, the biological consequences of these substances are not well understood. Identifying shared targets will inform cessation pharmacotherapies and provide a deeper understanding of how co-use of alcohol and nicotine impacts health, including biomarkers of stress and inflammation.Objective: We examined the effects of nicotine exposure and withdrawal on alcohol self-administration (SA), stress and inflammatory biomarkers, and a G-protein coupled receptor subunit (Gβ) in brain areas associated with drug use.Methods: Male rats were trained to SA alcohol and then received a nicotine pump (n = 7-8 per group). We assessed alcohol intake for 12 days during nicotine exposure and then following pump removal to elicit withdrawal. After the behavioral studies, we assessed plasma leptin, corticosterone, and interleukin-1β (IL-1β), and Gβ protein expression in the amygdala, nucleus accumbens (NAc), and prefrontal cortex (PFC).Results: Nicotine exposure or withdrawal did not alter alcohol intake (p > .05). Alcohol and nicotine withdrawal elevated corticosterone levels (p = .015) and decreased Gβ levels in the PFC (p = .004). In the absence of nicotine, alcohol SA suppressed IL-1β levels (p = .039). Chronic exposure to nicotine or withdrawal during alcohol SA did not alter leptin levels or Gβ expression in the amygdala or NAc (p's > .05).Conclusions: The combination of alcohol SA and nicotine withdrawal produced a persistent increase in stress biomarkers and a suppression in Gβ expression in the PFC, providing an important first step toward understanding the common biological mechanisms of alcohol/nicotine misuse.
Collapse
Affiliation(s)
- Bryan Cruz
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, USA
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Karen Castañeda
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA
| | - Michelle Aranda
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA
| | - Cecilia A. Hinojosa
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, USA
| | | | - Rodolfo J. Flores
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, USA
| | - Charles T. Spencer
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA
| | - Valentina Vozella
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Bharathi S. Gadad
- Department of Psychiatry, Paul L Foster School of Medicine, Texas Tech University Health Science Center, El Paso, TX, USA
- Southwest Brain Bank, Texas Tech University Health Science Center, El Paso, TX, USA
| | - Sukla Roychowdhury
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, USA
| | - Laura E. O’Dell
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, USA
| |
Collapse
|
7
|
Meqbil YJ, van Rijn RM. Opportunities and Challenges for In Silico Drug Discovery at Delta Opioid Receptors. Pharmaceuticals (Basel) 2022; 15:873. [PMID: 35890173 PMCID: PMC9324648 DOI: 10.3390/ph15070873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/13/2022] [Indexed: 11/29/2022] Open
Abstract
The delta opioid receptor is a Gi-protein-coupled receptor (GPCR) with a broad expression pattern both in the central nervous system and the body. The receptor has been investigated as a potential target for a multitude of significant diseases including migraine, alcohol use disorder, ischemia, and neurodegenerative diseases. Despite multiple attempts, delta opioid receptor-selective molecules have not been translated into the clinic. Yet, the therapeutic promise of the delta opioid receptor remains and thus there is a need to identify novel delta opioid receptor ligands to be optimized and selected for clinical trials. Here, we highlight recent developments involving the delta opioid receptor, the closely related mu and kappa opioid receptors, and in the broader area of the GPCR drug discovery research. We focus on the validity and utility of the available delta opioid receptor structures. We also discuss the increased ability to perform ultra-large-scale docking studies on GPCRs, the rise in high-resolution cryo-EM structures, and the increased prevalence of machine learning and artificial intelligence in drug discovery. Overall, we pose that there are multiple opportunities to enable in silico drug discovery at the delta opioid receptor to identify novel delta opioid modulators potentially with unique pharmacological properties, such as biased signaling.
Collapse
Affiliation(s)
- Yazan J. Meqbil
- Department of Medicinal Chemistry and Molecular Pharmacology, Computational Interdisciplinary Graduate Program, Purdue University, West Lafayette, IN 47907, USA;
| | - Richard M. van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue Institute for Neuroscience, Purdue University, West Lafayette, IN 47907, USA
- Septerna Inc., South San Francisco, CA 94080, USA
| |
Collapse
|
8
|
Chemogenetics as a neuromodulatory approach to treating neuropsychiatric diseases and disorders. Mol Ther 2022; 30:990-1005. [PMID: 34861415 PMCID: PMC8899595 DOI: 10.1016/j.ymthe.2021.11.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/12/2021] [Accepted: 11/29/2021] [Indexed: 01/01/2023] Open
Abstract
Chemogenetics enables precise, non-invasive, and reversible modulation of neural activity via the activation of engineered receptors that are pharmacologically selective to endogenous or exogenous ligands. With recent advances in therapeutic gene delivery, chemogenetics is poised to support novel interventions against neuropsychiatric diseases and disorders. To evaluate its translational potential, we performed a scoping review of applications of chemogenetics that led to the reversal of molecular and behavioral deficits in studies relevant to neuropsychiatric diseases and disorders. In this review, we present these findings and discuss the potential and challenges for using chemogenetics as a precision medicine-based neuromodulation strategy.
Collapse
|
9
|
Characterization of DREADD receptor expression and function in rhesus macaques trained to discriminate ethanol. Neuropsychopharmacology 2022; 47:857-865. [PMID: 34654906 PMCID: PMC8882175 DOI: 10.1038/s41386-021-01181-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 08/10/2021] [Accepted: 09/03/2021] [Indexed: 12/11/2022]
Abstract
Circuit manipulation has been a staple technique in neuroscience to identify how the brain functions to control complex behaviors. Chemogenetics, including designer receptors exclusively activated by designer drugs (DREADDs), have proven to be a powerful tool for the reversible modulation of discrete brain circuitry without the need for implantable devices, thereby making them especially useful in awake and unrestrained animals. This study used a DREADD approach to query the role of the nucleus accumbens (NAc) in mediating the interoceptive effects of 1.0 g/kg ethanol (i.g.) in rhesus monkeys (n = 7) using a drug discrimination procedure. After training, stereotaxic surgery was performed to introduce an AAV carrying the human muscarinic 4 receptor DREADD (hM4Di) bilaterally into the NAc. The hypothesis was that decreasing the output of the NAc by activation of hM4Di with the DREADD actuator, clozapine-n-oxide (CNO), would potentiate the discriminative stimulus effect of ethanol (i.e., a leftward shift the ethanol dose discrimination curve). The results showed individual variability shifts of the ethanol dose-response determination under DREADD activation. Characterization of the expression and function of hM4Di with MRI, immunohistochemical, and electrophysiological techniques found the selectivity of NAc transduction was proportional to behavioral effect. Specifically, the proportion of hM4Di expression restricted to the NAc was associated with the potency of the discriminative stimulus effects of ethanol. Together, these experiments highlight the NAc in mediating the interoceptive effects of ethanol, provide a framework for validation of chemogenetic tools in primates, and underscore the importance of robust within-subjects examination of DREADD expression for interpretation of behavioral findings.
Collapse
|
10
|
Mercuri NB, Federici M, Rizzo FR, Maugeri L, D'Addario SL, Ventura R, Berretta N. Long-Term Depression of Striatal DA Release Induced by mGluRs via Sustained Hyperactivity of Local Cholinergic Interneurons. Front Cell Neurosci 2021; 15:798464. [PMID: 34924961 PMCID: PMC8674918 DOI: 10.3389/fncel.2021.798464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 11/15/2021] [Indexed: 12/21/2022] Open
Abstract
The cellular mechanisms regulating dopamine (DA) release in the striatum have attracted much interest in recent years. By in vitro amperometric recordings in mouse striatal slices, we show that a brief (5 min) exposure to the metabotropic glutamate receptor agonist DHPG (50 μM) induces a profound depression of synaptic DA release, lasting over 1 h from DHPG washout. This long-term depression is sensitive to glycine, which preferentially inhibits local cholinergic interneurons, as well as to drugs acting on nicotinic acetylcholine receptors and to the pharmacological depletion of released acetylcholine. The same DHPG treatment induces a parallel long-lasting enhancement in the tonic firing of presumed striatal cholinergic interneurons, measured with multi-electrode array recordings. When DHPG is bilaterally infused in vivo in the mouse striatum, treated mice display an anxiety-like behavior. Our results demonstrate that metabotropic glutamate receptors stimulation gives rise to a prolonged depression of the striatal dopaminergic transmission, through a sustained enhancement of released acetylcholine, due to the parallel long-lasting potentiation of striatal cholinergic interneurons firing. This plastic interplay between dopamine, acetylcholine, and glutamate in the dorsal striatum may be involved in anxiety-like behavior typical of several neuropsychiatric disorders.
Collapse
Affiliation(s)
- Nicola B Mercuri
- IRCCS Fondazione Santa Lucia, Laboratory of Experimental Neurology, Rome, Italy.,Department of Systems Medicine, University of Tor Vergata, Rome, Italy
| | - Mauro Federici
- IRCCS Fondazione Santa Lucia, Laboratory of Experimental Neurology, Rome, Italy
| | | | - Lorenzo Maugeri
- IRCCS Fondazione Santa Lucia, Laboratory of Experimental Neurology, Rome, Italy
| | - Sebastian L D'Addario
- IRCCS Fondazione Santa Lucia, Laboratory of Experimental Neurology, Rome, Italy.,Department of Psychology and Center Daniel Bovet, Sapienza University, Rome, Italy.,Behavioral Neuroscience PhD Programme, Sapienza University, Rome, Italy
| | - Rossella Ventura
- IRCCS Fondazione Santa Lucia, Laboratory of Experimental Neurology, Rome, Italy.,Department of Psychology and Center Daniel Bovet, Sapienza University, Rome, Italy
| | - Nicola Berretta
- IRCCS Fondazione Santa Lucia, Laboratory of Experimental Neurology, Rome, Italy
| |
Collapse
|
11
|
Gutridge AM, Chakraborty S, Varga BR, Rhoda ES, French AR, Blaine AT, Royer QH, Cui H, Yuan J, Cassell RJ, Szabó M, Majumdar S, van Rijn RM. Evaluation of Kratom Opioid Derivatives as Potential Treatment Option for Alcohol Use Disorder. Front Pharmacol 2021; 12:764885. [PMID: 34803709 PMCID: PMC8596301 DOI: 10.3389/fphar.2021.764885] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022] Open
Abstract
Background and Purpose:Mitragyna speciosa extract and kratom alkaloids decrease alcohol consumption in mice at least in part through actions at the δ-opioid receptor (δOR). However, the most potent opioidergic kratom alkaloid, 7-hydroxymitragynine, exhibits rewarding properties and hyperlocomotion presumably due to preferred affinity for the mu opioid receptor (µOR). We hypothesized that opioidergic kratom alkaloids like paynantheine and speciogynine with reduced µOR potency could provide a starting point for developing opioids with an improved therapeutic window to treat alcohol use disorder. Experimental Approach: We characterized paynantheine, speciociliatine, and four novel kratom-derived analogs for their ability to bind and activate δOR, µOR, and κOR. Select opioids were assessed in behavioral assays in male C57BL/6N WT and δOR knockout mice. Key Results: Paynantheine (10 mg∙kg−1, i.p.) produced aversion in a limited conditioned place preference (CPP) paradigm but did not produce CPP with additional conditioning sessions. Paynantheine did not produce robust antinociception but did block morphine-induced antinociception and hyperlocomotion. Yet, at 10 and 30 mg∙kg−1 doses (i.p.), paynantheine did not counteract morphine CPP. 7-hydroxypaynantheine and 7-hydroxyspeciogynine displayed potency at δOR but limited µOR potency relative to 7-hydroxymitragynine in vitro, and dose-dependently decreased voluntary alcohol consumption in WT but not δOR in KO mice. 7-hydroxyspeciogynine has a maximally tolerated dose of at least 10 mg∙kg−1 (s.c.) at which it did not produce significant CPP neither alter general locomotion nor induce noticeable seizures. Conclusion and Implications: Derivatizing kratom alkaloids with the goal of enhancing δOR potency and reducing off-target effects could provide a pathway to develop novel lead compounds to treat alcohol use disorder with an improved therapeutic window.
Collapse
Affiliation(s)
- Anna M Gutridge
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Soumen Chakraborty
- Center for Clinical Pharmacology, University of Heath Sciences and Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO, United States
| | - Balazs R Varga
- Center for Clinical Pharmacology, University of Heath Sciences and Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO, United States
| | - Elizabeth S Rhoda
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Alexander R French
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States.,Purdue Institute for Integrative Neuroscience, West Lafayette, IN, United States
| | - Arryn T Blaine
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Quinten H Royer
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Haoyue Cui
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Jinling Yuan
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Robert J Cassell
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States.,Purdue Institute for Drug Discovery, West Lafayette, IN, United States
| | | | - Susruta Majumdar
- Center for Clinical Pharmacology, University of Heath Sciences and Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO, United States
| | - Richard M van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States.,Purdue Institute for Integrative Neuroscience, West Lafayette, IN, United States.,Purdue Institute for Drug Discovery, West Lafayette, IN, United States
| |
Collapse
|
12
|
Ko MJ, Chiang T, Mukadam AA, Mulia GE, Gutridge AM, Lin A, Chester JA, van Rijn RM. β-Arrestin-dependent ERK signaling reduces anxiety-like and conditioned fear-related behaviors in mice. Sci Signal 2021; 14:14/694/eaba0245. [PMID: 34344831 DOI: 10.1126/scisignal.aba0245] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
G protein-coupled receptors (GPCRs) are implicated in the regulation of fear and anxiety. GPCR signaling involves canonical G protein pathways but can also engage downstream kinases and effectors through scaffolding interactions mediated by β-arrestin. Here, we investigated whether β-arrestin signaling regulates anxiety-like and fear-related behavior in mice in response to activation of the GPCR δ-opioid receptor (δOR or DOR). Administration of β-arrestin-biased δOR agonists to male C57BL/6 mice revealed β-arrestin 2-dependent activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) in the dorsal hippocampus and amygdala and β-arrestin 1-dependent activation of ERK1/2 in the nucleus accumbens. In mice, β-arrestin-biased agonist treatment was associated with reduced anxiety-like and fear-related behaviors, with some overlapping and isoform-specific input. In contrast, applying a G protein-biased δOR agonist decreased ERK1/2 activity in all three regions as well as the dorsal striatum and was associated with increased fear-related behavior without effects on baseline anxiety. Our results indicate a complex picture of δOR neuromodulation in which β-arrestin 1- and 2-dependent ERK signaling in specific brain subregions suppresses behaviors associated with anxiety and fear and opposes the effects of G protein-biased signaling. Overall, our findings highlight the importance of noncanonical β-arrestin-dependent GPCR signaling in the regulation of these interrelated emotions.
Collapse
Affiliation(s)
- Mee Jung Ko
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA.,Purdue Institute for Integrative Neuroscience, West Lafayette, IN 47907, USA.,Purdue Interdisciplinary Life Sciences Graduate Program, West Lafayette, IN 47907, USA
| | - Terrance Chiang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Arbaaz A Mukadam
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA.,Department of Psychological Sciences, College of Health and Human Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Grace E Mulia
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA.,Purdue Interdisciplinary Life Sciences Graduate Program, West Lafayette, IN 47907, USA
| | - Anna M Gutridge
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA.,Purdue Institute for Integrative Neuroscience, West Lafayette, IN 47907, USA
| | - Angel Lin
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Julia A Chester
- Purdue Institute for Integrative Neuroscience, West Lafayette, IN 47907, USA.,Purdue Interdisciplinary Life Sciences Graduate Program, West Lafayette, IN 47907, USA.,Department of Psychological Sciences, College of Health and Human Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Richard M van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA. .,Purdue Institute for Integrative Neuroscience, West Lafayette, IN 47907, USA.,Purdue Interdisciplinary Life Sciences Graduate Program, West Lafayette, IN 47907, USA.,Purdue Institute for Drug Discovery, West Lafayette, IN 47907, USA
| |
Collapse
|
13
|
De Neve J, Barlow TMA, Tourwé D, Bihel F, Simonin F, Ballet S. Comprehensive overview of biased pharmacology at the opioid receptors: biased ligands and bias factors. RSC Med Chem 2021; 12:828-870. [PMID: 34223156 PMCID: PMC8221262 DOI: 10.1039/d1md00041a] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 03/30/2021] [Indexed: 12/19/2022] Open
Abstract
One of the main challenges in contemporary medicinal chemistry is the development of safer analgesics, used in the treatment of pain. Currently, moderate to severe pain is still treated with the "gold standard" opioids whose long-term often leads to severe side effects. With the discovery of biased agonism, the importance of this area of pharmacology has grown exponentially over the past decade. Of these side effects, tolerance, opioid misuse, physical dependence and substance use disorder (SUD) stand out, since these have led to many deaths over the past decades in both USA and Europe. New therapeutic molecules that induce a biased response at the opioid receptors (MOR, DOR, KOR and NOP receptor) are able to circumvent these side effects and, consequently, serve as more advantageous therapies with great promise. The concept of biased signaling extends far beyond the already sizeable field of GPCR pharmacology and covering everything would be vastly outside the scope of this review which consequently covers the biased ligands acting at the opioid family of receptors. The limitation of quantifying bias, however, makes this a controversial subject, where it is dependent on the reference ligand, the equation or the assay used for the quantification. Hence, the major issue in the field of biased ligands remains the translation of the in vitro profiles of biased signaling, with corresponding bias factors to in vivo profiles showing the presence or the lack of specific side effects. This review comprises a comprehensive overview of biased ligands in addition to their bias factors at individual members of the opioid family of receptors, as well as bifunctional ligands.
Collapse
Affiliation(s)
- Jolien De Neve
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel Brussels Belgium
| | - Thomas M A Barlow
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel Brussels Belgium
| | - Dirk Tourwé
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel Brussels Belgium
| | - Frédéric Bihel
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200, CNRS Université de Strasbourg Illkirch France
| | - Frédéric Simonin
- Biotechnologie et Signalisation Cellulaire, UMR 7242, CNRS, Université de Strasbourg Illkirch France
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel Brussels Belgium
| |
Collapse
|
14
|
Abstract
This paper is the forty-first consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2018 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (2), the roles of these opioid peptides and receptors in pain and analgesia in animals (3) and humans (4), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (5), opioid peptide and receptor involvement in tolerance and dependence (6), stress and social status (7), learning and memory (8), eating and drinking (9), drug abuse and alcohol (10), sexual activity and hormones, pregnancy, development and endocrinology (11), mental illness and mood (12), seizures and neurologic disorders (13), electrical-related activity and neurophysiology (14), general activity and locomotion (15), gastrointestinal, renal and hepatic functions (16), cardiovascular responses (17), respiration and thermoregulation (18), and immunological responses (19).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY, 11367, United States.
| |
Collapse
|
15
|
Gutridge AM, Robins MT, Cassell RJ, Uprety R, Mores KL, Ko MJ, Pasternak GW, Majumdar S, van Rijn RM. G protein-biased kratom-alkaloids and synthetic carfentanil-amide opioids as potential treatments for alcohol use disorder. Br J Pharmacol 2020; 177:1497-1513. [PMID: 31705528 PMCID: PMC7060366 DOI: 10.1111/bph.14913] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/12/2019] [Accepted: 10/15/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Mitragyna speciosa, more commonly known as kratom, is a plant that contains opioidergic alkaloids but is unregulated in most countries. Kratom is used in the self-medication of chronic pain and to reduce illicit and prescription opioid dependence. Kratom may be less dangerous than typical opioids because of the stronger preference of kratom alkaloids to induce receptor interaction with G proteins over β-arrestin proteins. We hypothesized that kratom (alkaloids) can also reduce alcohol intake. EXPERIMENTAL APPROACH We pharmacologically characterized kratom extracts, kratom alkaloids (mitragynine, 7-hydroxymitragynine, paynantheine, and speciogynine) and synthetic carfentanil-amide opioids for their ability to interact with G proteins and β-arrestin at μ, δ, and κ opioid receptors in vitro. We used C57BL/6 mice to assess to which degree these opioids could reduce alcohol intake and whether they had rewarding properties. KEY RESULTS Kratom alkaloids were strongly G protein-biased at all three opioid receptors and reduced alcohol intake, but kratom and 7-hydroxymitragynine were rewarding. Several results indicated a key role for δ opioid receptors, including that the synthetic carfentanil-amide opioid MP102-a G protein-biased agonist with modest selectivity for δ opioid receptors-reduced alcohol intake, whereas the G protein-biased μ opioid agonist TRV130 did not. CONCLUSION AND IMPLICATIONS Our results suggest that kratom extracts can decrease alcohol intake but still carry significant risk upon prolonged use. Development of more δ opioid-selective synthetic opioids may provide a safer option than kratom to treat alcohol use disorder with fewer side effects.
Collapse
Affiliation(s)
- Anna M. Gutridge
- Department of Medicinal Chemistry and Molecular Pharmacology, College of PharmacyPurdue UniversityWest LafayetteIndiana
| | - Meridith T. Robins
- Department of Medicinal Chemistry and Molecular Pharmacology, College of PharmacyPurdue UniversityWest LafayetteIndiana
| | - Robert J. Cassell
- Department of Medicinal Chemistry and Molecular Pharmacology, College of PharmacyPurdue UniversityWest LafayetteIndiana
| | - Rajendra Uprety
- Department of Neurology and Molecular PharmacologyMemorial Sloan Kettering Cancer CenterNew YorkNew York
| | - Kendall L. Mores
- Department of Medicinal Chemistry and Molecular Pharmacology, College of PharmacyPurdue UniversityWest LafayetteIndiana
| | - Mee Jung Ko
- Department of Medicinal Chemistry and Molecular Pharmacology, College of PharmacyPurdue UniversityWest LafayetteIndiana
- Purdue Interdisciplinary Life Sciences Graduate ProgramPurdue UniversityWest LafayetteIndiana
| | - Gavril W. Pasternak
- Department of Neurology and Molecular PharmacologyMemorial Sloan Kettering Cancer CenterNew YorkNew York
| | - Susruta Majumdar
- Department of Neurology and Molecular PharmacologyMemorial Sloan Kettering Cancer CenterNew YorkNew York
- Center for Clinical PharmacologySt. Louis College of Pharmacy and Washington University School of MedicineSt. LouisMissouri
| | - Richard M. van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, College of PharmacyPurdue UniversityWest LafayetteIndiana
- Purdue Institute for Drug DiscoveryPurdue UniversityWest LafayetteIndiana
- Purdue Institute for Integrative NeurosciencePurdue UniversityWest LafayetteIndiana
- Purdue Interdisciplinary Life Sciences Graduate ProgramPurdue UniversityWest LafayetteIndiana
| |
Collapse
|
16
|
Cassell RJ, Sharma KK, Su H, Cummins BR, Cui H, Mores KL, Blaine AT, Altman RA, van Rijn RM. The Meta-Position of Phe 4 in Leu-Enkephalin Regulates Potency, Selectivity, Functional Activity, and Signaling Bias at the Delta and Mu Opioid Receptors. Molecules 2019; 24:molecules24244542. [PMID: 31842282 PMCID: PMC6943441 DOI: 10.3390/molecules24244542] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/08/2019] [Accepted: 12/10/2019] [Indexed: 01/10/2023] Open
Abstract
As tool compounds to study cardiac ischemia, the endogenous δ-opioid receptors (δOR) agonist Leu5-enkephalin and the more metabolically stable synthetic peptide (d-Ala2, d-Leu5)-enkephalin are frequently employed. However, both peptides have similar pharmacological profiles that restrict detailed investigation of the cellular mechanism of the δOR’s protective role during ischemic events. Thus, a need remains for δOR peptides with improved selectivity and unique signaling properties for investigating the specific roles for δOR signaling in cardiac ischemia. To this end, we explored substitution at the Phe4 position of Leu5-enkephalin for its ability to modulate receptor function and selectivity. Peptides were assessed for their affinity to bind to δORs and µ-opioid receptors (µORs) and potency to inhibit cAMP signaling and to recruit β-arrestin 2. Additionally, peptide stability was measured in rat plasma. Substitution of the meta-position of Phe4 of Leu5-enkephalin provided high-affinity ligands with varying levels of selectivity and bias at both the δOR and µOR and improved peptide stability, while substitution with picoline derivatives produced lower-affinity ligands with G protein biases at both receptors. Overall, these favorable substitutions at the meta-position of Phe4 may be combined with other modifications to Leu5-enkephalin to deliver improved agonists with finely tuned potency, selectivity, bias and drug-like properties.
Collapse
MESH Headings
- Animals
- CHO Cells
- Cricetulus
- Enkephalin, Leucine/genetics
- Enkephalin, Leucine/pharmacology
- Humans
- Phenylalanine
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
Collapse
Affiliation(s)
- Robert J. Cassell
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA; (R.J.C.); (H.S.); (K.L.M.); (A.T.B.)
| | - Krishna K. Sharma
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, KS 66045, USA;
| | - Hongyu Su
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA; (R.J.C.); (H.S.); (K.L.M.); (A.T.B.)
| | | | - Haoyue Cui
- College of Wuya, Shenyang Pharmaceutical University, Shenyang 110016, China;
| | - Kendall L. Mores
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA; (R.J.C.); (H.S.); (K.L.M.); (A.T.B.)
| | - Arryn T. Blaine
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA; (R.J.C.); (H.S.); (K.L.M.); (A.T.B.)
| | - Ryan A. Altman
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, KS 66045, USA;
- Correspondence: (R.A.A.); (R.M.v.R.)
| | - Richard M. van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA; (R.J.C.); (H.S.); (K.L.M.); (A.T.B.)
- Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
- Correspondence: (R.A.A.); (R.M.v.R.)
| |
Collapse
|
17
|
Mores KL, Cummins BR, Cassell RJ, van Rijn RM. A Review of the Therapeutic Potential of Recently Developed G Protein-Biased Kappa Agonists. Front Pharmacol 2019; 10:407. [PMID: 31057409 PMCID: PMC6478756 DOI: 10.3389/fphar.2019.00407] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 04/01/2019] [Indexed: 01/22/2023] Open
Abstract
Between 2000 and 2005 several studies revealed that morphine is more potent and exhibits fewer side effects in beta-arrestin 2 knockout mice. These findings spurred efforts to develop opioids that signal primarily via G protein activation and do not, or only very weakly, recruit beta-arrestin. Development of such molecules targeting the mu opioid receptor initially outpaced those targeting the kappa, delta and nociceptin opioid receptors, with the G protein-biased mu opioid agonist oliceridine/TRV130 having completed phase III clinical trials with improved therapeutic window to treat moderate-to-severe acute pain. Recently however, there has been a sharp increase in the development of novel G protein-biased kappa agonists. It is hypothesized that G protein-biased kappa agonists can reduce pain and itch, but exhibit fewer side effects, such as anhedonia and psychosis, that have thus far limited the clinical development of unbiased kappa opioid agonists. Here we summarize recently discovered G protein-biased kappa agonists, comparing structures, degree of signal bias and preclinical effects. We specifically reviewed nalfurafine, 22-thiocyanatosalvinorin A (RB-64), mesyl-salvinorin B, 2-(4-(furan-2-ylmethyl)-5-((4-methyl-3-(trifluoromethyl)benzyl)thio)-4H-1,2,4-triazol-3-yl)pyridine (triazole 1.1), 3-(2-((cyclopropylmethyl)(phenethyl)amino)ethyl)phenol (HS666), N-n-butyl-N-phenylethyl-N-3-hydroxyphenylethyl-amine (compound 5/BPHA), 6-guanidinonaltrindole (6′GNTI), and collybolide. These agonists encompass a variety of chemical scaffolds and range in both their potency and efficacy in terms of G protein signaling and beta-arrestin recruitment. Thus unsurprisingly, the behavioral responses reported for these agonists are not uniform. Yet, it is our conclusion that the kappa opioid field will benefit tremendously from future studies that compare several biased agonists and correlate the degree of signaling bias to a particular pharmacological response.
Collapse
Affiliation(s)
- Kendall L Mores
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, West Lafayette, IN, United States
| | - Benjamin R Cummins
- Department of Chemistry, College of Science, West Lafayette, IN, United States
| | - Robert J Cassell
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, West Lafayette, IN, United States.,Purdue Institute for Drug Discovery, West Lafayette, IN, United States
| | - Richard M van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, West Lafayette, IN, United States.,Purdue Institute for Drug Discovery, West Lafayette, IN, United States.,Purdue Institute for Integrative Neuroscience, West Lafayette, IN, United States
| |
Collapse
|
18
|
Cassell RJ, Mores KL, Zerfas BL, Mahmoud AH, Lill MA, Trader DJ, van Rijn RM. Rubiscolins are naturally occurring G protein-biased delta opioid receptor peptides. Eur Neuropsychopharmacol 2019; 29:450-456. [PMID: 30591345 PMCID: PMC6421079 DOI: 10.1016/j.euroneuro.2018.12.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/19/2018] [Accepted: 12/16/2018] [Indexed: 10/27/2022]
Abstract
The impact that β-arrestin proteins have on G protein-coupled receptor trafficking, signaling and physiological behavior has gained much appreciation over the past decade. A number of studies have attributed the side effects associated with the use of naturally occurring and synthetic opioids, such as respiratory depression and constipation, to excessive recruitment of β-arrestin. These findings have led to the development of biased opioid small molecule agonists that do not recruit β-arrestin, activating only the canonical G protein pathway. Similar G protein-biased small molecule opioids have been found to occur in nature, particularly within kratom, and opioids within salvia have served as a template for the synthesis of other G protein-biased opioids. Here, we present the first report of naturally occurring peptides that selectively activate G protein signaling pathways at δ opioid receptors, but with minimal β-arrestin recruitment. Specifically, we find that rubiscolin peptides, which are produced as cleavage products of the plant protein rubisco, bind to and activate G protein signaling at δ opioid receptors. However, unlike the naturally occurring δ opioid peptides leu-enkephalin and deltorphin II, the rubiscolin peptides only very weakly recruit β-arrestin 2 and have undetectable recruitment of β-arrestin 1 at the δ opioid receptor.
Collapse
Affiliation(s)
- Robert J Cassell
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Kendall L Mores
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Breanna L Zerfas
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Amr H Mahmoud
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Markus A Lill
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, West Lafayette, IN 47907, USA; Purdue Institute for Integrative Neuroscience, West Lafayette, IN 47907, USA
| | - Darci J Trader
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, West Lafayette, IN 47907, USA; Purdue Institute for Integrative Neuroscience, West Lafayette, IN 47907, USA
| | - Richard M van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, West Lafayette, IN 47907, USA; Purdue Institute for Integrative Neuroscience, West Lafayette, IN 47907, USA.
| |
Collapse
|
19
|
Mores KL, Cassell RJ, van Rijn RM. Arrestin recruitment and signaling by G protein-coupled receptor heteromers. Neuropharmacology 2018; 152:15-21. [PMID: 30419245 DOI: 10.1016/j.neuropharm.2018.11.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/28/2018] [Accepted: 11/08/2018] [Indexed: 12/12/2022]
Abstract
G protein-coupled receptors (GPCR) have a long history of being considered a prime target for drug development to treat a plethora of diseases and disorders. In fact in 1827, the first approved therapeutic in the United States was morphine, a drug that targets a GPCR, namely the mu opioid receptor. However, with the rise in biologics over the last two decades, the market share of small molecules targeting GPCRs has declined. Still, two phenomena concerning GPCR pharmacology, specifically heteromerization and biased signaling, have bolstered new interests in this particular class of drug targets. Heteromerization, the process by which two distinct GPCRs come together to form a unique signaling complex, has been demonstrated between many different GPCRs and has spurred efforts to discover heteromer selective drugs. Additionally, the discovery of biased signaling, a concept by which a GPCR can transduce intracellular signaling by favoring a specific pathway (e.g. G-protein) over another pathway (e.g. arrestin), has led to the development of signal-biased drugs with potentially fewer side effects. Our goal for this review is to highlight studies that have investigated the interplay of these two phenomena by providing an overview of the current literature describing instances where GPCR heteromers have distinct arrestin recruitment profiles when compared to the individual GPCRs, with a focus on those GPCRs expressed in the central nervous system. This article is part of the Special Issue entitled 'Receptor heteromers and their allosteric receptor-receptor interactions'.
Collapse
Affiliation(s)
- Kendall L Mores
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, United States
| | - Robert J Cassell
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, United States
| | - Richard M van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, United States; Purdue Institute for Drug Discovery, United States; Purdue Institute for Integrative Neuroscience, West Lafayette, IN, 47907, United States.
| |
Collapse
|
20
|
Alper K, Dong B, Shah R, Sershen H, Vinod KY. LSD Administered as a Single Dose Reduces Alcohol Consumption in C57BL/6J Mice. Front Pharmacol 2018; 9:994. [PMID: 30233372 PMCID: PMC6127266 DOI: 10.3389/fphar.2018.00994] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 08/13/2018] [Indexed: 12/31/2022] Open
Abstract
There is a substantive clinical literature on classical hallucinogens, most commonly lysergic acid diethylamide (LSD) for the treatment of alcohol use disorder. However, there has been no published research on the effect of LSD on alcohol consumption in animals. This study evaluated the effect of LSD in mice using a two-bottle choice alcohol drinking paradigm. Adult male C57BL/6J mice were exposed to ethanol to develop preference and divided into three groups of equal ethanol consumption, and then treated with single intraperitoneal injection of saline or 25 or 50 μg/kg LSD and offered water and 20% ethanol. The respective LSD-treated groups were compared to the control group utilizing a multilevel model for repeated measures. In mice treated with 50 μg/kg LSD ethanol consumption was reduced relative to controls (p = 0.0035), as was ethanol preference (p = 0.0024), with a group mean reduction of ethanol consumption of 17.9% sustained over an interval of 46 days following LSD administration. No significant effects on ethanol consumption or preference were observed in mice treated with 25 μg/kg LSD. Neither total fluid intake nor locomotor activity in the LSD-treated groups differed significantly from controls. These results suggest that classical hallucinogens in the animal model merit further study as a potential approach to the identification of targets for drug discovery and investigation of the neurobiology of addiction.
Collapse
Affiliation(s)
- Kenneth Alper
- Department of Psychiatry, New York University School of Medicine, New York, NY, United States.,Department of Neurology, New York University School of Medicine, New York, NY, United States
| | - Bin Dong
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - Relish Shah
- Emotional Brain Institute, Orangeburg, NY, United States
| | - Henry Sershen
- Department of Psychiatry, New York University School of Medicine, New York, NY, United States.,Department of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - K Yaragudri Vinod
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States.,Emotional Brain Institute, Orangeburg, NY, United States.,Department of Child and Adolescent Psychiatry, New York University School of Medicine, New York, NY, United States
| |
Collapse
|