1
|
Hiranita T, Hong WC, Sharma A, Lopez JP, Mesangeau C, Whittaker DA, Alsharif W, Kopajtic TA, Jamalapuram S, Avery BA, Tanda G, McCurdy CR, Katz JL. Preclinical Profile of CM699 as a Medication Candidate for Stimulant Use Disorder. ACS Chem Neurosci 2025; 16:1454-1468. [PMID: 40132017 DOI: 10.1021/acschemneuro.4c00589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025] Open
Abstract
There currently are no medications proven to be effective for the treatment of stimulant-use disorder (SUD). Sigma-receptor (σR) antagonists block many effects of stimulant drugs but not the reinforcing effects assessed with self-administration in rats. However, a recent study suggests that σR antagonism combined with a dopamine (DA) transporter (DAT) blockade selectively attenuates stimulant self-administration. A compound with potential for dual DAT/σR inhibition, CM699, was synthesized and had the necessary ex vivo affinities of 311 and 14.1 nM at DAT and σ1Rs, respectively. CM699 inhibited DA uptake ex vivo. Antagonist effects at σ1Rs by CM699 were confirmed with a recently reported pharmacological assay: CM699 increased, whereas the σ1R agonist, (+)-pentazocine, decreased σ1R multimers detected in nondenaturing protein gels, and CM699 blocked the effects of (+)-pentazocine. CM699 after intravenous administration (5.0 mg/kg) in rats had an elimination half-life of 4.4 h. In rats, CM699 after intraperitoneal administration blunted the stimulatory effects of cocaine on DA levels in the nucleus accumbens and insurmountably blocked cocaine self-administration, indicating efficacy as a cocaine antagonist in vivo. When given alone, CM699 was not self-administered nor had significant effects on nucleus accumbens DA, suggesting minimal, if any, abuse potential. Further, in a biochemical assay designed to probe the conformation of DAT, (+)-pentazocine potentiated cocaine-induced cysteine accessibility of DAT transmembrane domain 6a, suggesting a shift in the conformational equilibrium of DAT toward outward-facing, whereas CM699 blocked this effect. The results provide preclinical proof of concept for dual DAT/σR inhibition as a novel DAT-conformational approach for the development of medications to treat SUD.
Collapse
Affiliation(s)
- Takato Hiranita
- Department of Pharmacology, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas 78229, United States
| | - Weimin C Hong
- Department of Pharmaceutical Sciences, Butler University, Indianapolis, Indiana 46208, United States
| | - Abhisheak Sharma
- Department of Pharmaceutics, University of Florida, Gainesville, Florida 32610, United States
- Translational Drug Development Core, University of Florida, Gainesville, Florida 32610, United States
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, University Mississippi 38677, United States
| | - Jessica P Lopez
- Psychobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Christophe Mesangeau
- Department Biomolecular Sciences, University of Mississippi, University, Mississippi 38677, United States
| | - Daniel A Whittaker
- Department of Pharmaceutical Sciences, Butler University, Indianapolis, Indiana 46208, United States
| | - Walid Alsharif
- Department Biomolecular Sciences, University of Mississippi, University, Mississippi 38677, United States
| | - Theresa A Kopajtic
- Psychobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Seshulatha Jamalapuram
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, University Mississippi 38677, United States
| | - Bonnie A Avery
- Department of Pharmaceutics, University of Florida, Gainesville, Florida 32610, United States
- Translational Drug Development Core, University of Florida, Gainesville, Florida 32610, United States
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, University Mississippi 38677, United States
| | - Gianluigi Tanda
- Medication Development Program, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Christopher R McCurdy
- Department of Pharmaceutics, University of Florida, Gainesville, Florida 32610, United States
- Translational Drug Development Core, University of Florida, Gainesville, Florida 32610, United States
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, University Mississippi 38677, United States
| | - Jonathan L Katz
- Psychobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224, United States
| |
Collapse
|
2
|
Grimm AK, Rozanes D, Shpigel E, Moscovici L, Belkin S. A Microbial Cocaine Bioreporter. SENSORS (BASEL, SWITZERLAND) 2024; 24:6549. [PMID: 39460031 PMCID: PMC11511522 DOI: 10.3390/s24206549] [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: 09/01/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024]
Abstract
The continuous emergence of new illegal compounds, particularly psychoactive chemicals, poses significant challenges for current drug detection methods. Developing new protocols and kits for each new drug requires substantial time, effort, and dedicated manpower. Whole-cell bacterial bioreporters have been proven capable of detecting diverse hazardous compounds in both laboratory and field settings, identifying not only single compounds but also chemical families. We present the development of a microbial bioreporter for the detection of cocaine, the nervous system stimulant that is the second-most widely used illegal drug in the US. Escherichia coli was transformed with a plasmid containing a bacterial luxCDABEG bioluminescence gene cassette, activated by a cocaine-responsive signaling cascade. The engineered bioreporter is demonstrated to be a sensitive and specific first-generation detection system for cocaine, with detection thresholds of 17 ± 8 μg/L and 130 ± 50 μg/L in a buffer solution and in urine, respectively. Further improvement of the sensor's performance was achieved by altering the nucleotide sequence of the PBen gene promoter, the construct's sensing element, using accelerated site-directed evolution. The applicability of ready-to-use paper strips with immobilized bioreporter cells was demonstrated for cocaine detection in aqueous solutions.
Collapse
Affiliation(s)
- Anne-Kathrin Grimm
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, 93053 Regensburg, Germany;
| | - Dor Rozanes
- Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; (D.R.); (E.S.); (L.M.)
| | - Etai Shpigel
- Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; (D.R.); (E.S.); (L.M.)
| | - Liat Moscovici
- Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; (D.R.); (E.S.); (L.M.)
| | - Shimshon Belkin
- Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; (D.R.); (E.S.); (L.M.)
| |
Collapse
|
3
|
Mu L, Liu X, Yu H, Vickstrom CR, Friedman V, Kelly TJ, Hu Y, Su W, Liu S, Mantsch JR, Liu QS. cAMP-mediated upregulation of HCN channels in VTA dopamine neurons promotes cocaine reinforcement. Mol Psychiatry 2023; 28:3930-3942. [PMID: 37845497 PMCID: PMC10730389 DOI: 10.1038/s41380-023-02290-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 10/18/2023]
Abstract
Chronic cocaine exposure induces enduring neuroadaptations that facilitate motivated drug taking. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are known to modulate neuronal firing and pacemaker activity in ventral tegmental area (VTA) dopamine neurons. However, it remained unknown whether cocaine self-administration affects HCN channel function and whether HCN channel activity modulates motivated drug taking. We report that rat VTA dopamine neurons predominantly express Hcn3-4 mRNA, while VTA GABA neurons express Hcn1-4 mRNA. Both neuronal types display similar hyperpolarization-activated currents (Ih), which are facilitated by acute increases in cAMP. Acute cocaine application decreases voltage-dependent activation of Ih in VTA dopamine neurons, but not in GABA neurons. Unexpectedly, chronic cocaine self-administration results in enhanced Ih selectively in VTA dopamine neurons. This differential modulation of Ih currents is likely mediated by a D2 autoreceptor-induced decrease in cAMP as D2 (Drd2) mRNA is predominantly expressed in dopamine neurons, whereas D1 (Drd1) mRNA is barely detectable in the VTA. Moreover, chronically decreased cAMP via Gi-DREADD stimulation leads to an increase in Ih in VTA dopamine neurons and enhanced binding of HCN3/HCN4 with tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b), an auxiliary subunit that is known to facilitate HCN channel surface trafficking. Finally, we show that systemic injection and intra-VTA infusion of the HCN blocker ivabradine reduces cocaine self-administration under a progressive ratio schedule and produces a downward shift of the cocaine dose-response curve. Our results suggest that cocaine self-administration induces an upregulation of Ih in VTA dopamine neurons, while HCN inhibition reduces the motivation for cocaine intake.
Collapse
Affiliation(s)
- Lianwei Mu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Xiaojie Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Hao Yu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Casey R Vickstrom
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Vladislav Friedman
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Thomas J Kelly
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Ying Hu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Wantang Su
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Department of Exercise Physiology, Beijing Sport University, Beijing, 100084, China
| | - Shuai Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - John R Mantsch
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
4
|
Hersey M, Bartole MK, Jones CS, Newman AH, Tanda G. Are There Prevalent Sex Differences in Psychostimulant Use Disorder? A Focus on the Potential Therapeutic Efficacy of Atypical Dopamine Uptake Inhibitors. Molecules 2023; 28:5270. [PMID: 37446929 PMCID: PMC10343811 DOI: 10.3390/molecules28135270] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Psychostimulant use disorders (PSUD) affect a growing number of men and women and exert sizable public health and economic burdens on our global society. Notably, there are some sex differences in the onset of dependence, relapse rates, and treatment success with PSUD observed in preclinical and clinical studies. The subtle sex differences observed in the behavioral aspects of PSUD may be associated with differences in the neurochemistry of the dopaminergic system between sexes. Preclinically, psychostimulants have been shown to increase synaptic dopamine (DA) levels and may downregulate the dopamine transporter (DAT). This effect is greatest in females during the high estradiol phase of the estrous cycle. Interestingly, women have been shown to be more likely to begin drug use at younger ages and report higher levels of desire to use cocaine than males. Even though there is currently no FDA-approved medication, modafinil, a DAT inhibitor approved for use in the treatment of narcolepsy and sleep disorders, has shown promise in the treatment of PSUD among specific populations of affected individuals. In this review, we highlight the therapeutic potential of modafinil and other atypical DAT inhibitors focusing on the lack of sex differences in the actions of these agents.
Collapse
Affiliation(s)
| | | | | | | | - Gianluigi Tanda
- Medication Development Program, NIDA IRP, Baltimore, MD 21224, USA; (M.H.); (M.K.B.); (C.S.J.); (A.H.N.)
| |
Collapse
|
5
|
Keighron JD, Bonaventura J, Li Y, Yang JW, DeMarco EM, Hersey M, Cao J, Sandtner W, Michaelides M, Sitte HH, Newman AH, Tanda G. Interactions of calmodulin kinase II with the dopamine transporter facilitate cocaine-induced enhancement of evoked dopamine release. Transl Psychiatry 2023; 13:202. [PMID: 37311803 PMCID: PMC10264427 DOI: 10.1038/s41398-023-02493-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/17/2023] [Accepted: 05/26/2023] [Indexed: 06/15/2023] Open
Abstract
Typical and atypical dopamine uptake inhibitors (DUIs) prefer distinct conformations of the dopamine transporter (DAT) to form ligand-transporter complexes, resulting in markedly different effects on behavior, neurochemistry, and potential for addiction. Here we show that cocaine and cocaine-like typical psychostimulants elicit changes in DA dynamics distinct from those elicited by atypical DUIs, as measured via voltammetry procedures. While both classes of DUIs reduced DA clearance rate, an effect significantly related to their DAT affinity, only typical DUIs elicited a significant stimulation of evoked DA release, an effect unrelated to their DAT affinity, which suggests a mechanism of action other than or in addition to DAT blockade. When given in combination, typical DUIs enhance the stimulatory effects of cocaine on evoked DA release while atypical DUIs blunt them. Pretreatments with an inhibitor of CaMKIIα, a kinase that interacts with DAT and that regulates synapsin phosphorylation and mobilization of reserve pools of DA vesicles, blunted the effects of cocaine on evoked DA release. Our results suggest a role for CaMKIIα in modulating the effects of cocaine on evoked DA release without affecting cocaine inhibition of DA reuptake. This effect is related to a specific DAT conformation stabilized by cocaine. Moreover, atypical DUIs, which prefer a distinct DAT conformation, blunt cocaine's neurochemical and behavioral effects, indicating a unique mechanism underlying their potential as medications for treating psychostimulant use disorder.
Collapse
Affiliation(s)
- Jacqueline D Keighron
- Medication Development Program, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
- Department of Biological and Chemical Science, New York Institute of Technology, Old Westbury, NY, USA
| | - Jordi Bonaventura
- Biobehavioral Imaging & Molecular Neuropsychopharmacology Unit, Neuroimaging Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
- Department of Pathology and Experimental Therapeutics, Institut de Neurociències, Universitat de Barcelona, L'Hospitalet de Llobregat, Catalonia, Spain
| | - Yang Li
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jae-Won Yang
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Emily M DeMarco
- Medication Development Program, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Melinda Hersey
- Medication Development Program, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Jianjing Cao
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Walter Sandtner
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael Michaelides
- Biobehavioral Imaging & Molecular Neuropsychopharmacology Unit, Neuroimaging Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Harald H Sitte
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Amy Hauck Newman
- Medication Development Program, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Gianluigi Tanda
- Medication Development Program, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA.
| |
Collapse
|
6
|
Rahimi O, Cao J, Lam J, Childers SR, Rais R, Porrino LJ, Newman AH, Nader MA. The Effects of the Dopamine Transporter Ligands JJC8-088 and JJC8-091 on Cocaine versus Food Choice in Rhesus Monkeys. J Pharmacol Exp Ther 2023; 384:372-381. [PMID: 36507847 PMCID: PMC9976790 DOI: 10.1124/jpet.122.001363] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 11/26/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Although there are no Food and Drug Administration-approved treatments for cocaine use disorder, several modafinil analogs have demonstrated promise in reducing cocaine self-administration and reinstatement in rats. Furthermore, the range of dopamine transporter (DAT) compounds provides an opportunity to develop pharmacotherapeutics without abuse liability. This study extended the comparison of JJC8-088 and JJC8-091, the former compound having higher DAT affinity and predicted abuse liability, to rhesus monkeys using a concurrent cocaine versus food schedule of reinforcement. First, binding to striatal DAT was examined in cocaine-naïve monkey tissue. Next, intravenous pharmacokinetics of both JJC compounds were evaluated in cocaine-experienced male monkeys (n = 3/drug). In behavioral studies, acute and chronic administration of both compounds were evaluated in these same monkeys responding under a concurrent food versus cocaine (0 and 0.003-0.1 mg/kg per injection) schedule of reinforcement. In nonhuman primate striatum, JJC8-088 had higher DAT affinity compared with JJC8-091 (14.4 ± 9 versus 2730 ± 1270 nM, respectively). Both JJC compounds had favorable plasma pharmacokinetics for behavioral assessments, with half-lives of 1.1 hours and 3.5 hours for JJC8-088 (0.7 mg/kg, i.v.) and JJC8-091 (1.9 mg/kg, i.v.), respectively. Acute treatment with both compounds shifted the cocaine dose-response curve to the left. Chronic treatment with JJC8-088 decreased cocaine choice in two of the three monkeys, whereas JJC8-091 only modestly reduced cocaine allocation in one monkey. Differences in affinities of JJC8-091 DAT binding in monkeys compared with rats may account for the poor rodent-to-monkey translation. Future studies should evaluate atypical DAT blockers in combination with behavioral interventions that may further decrease cocaine choice. SIGNIFICANCE STATEMENT: Cocaine use disorder (CUD) remains a significant public health problem with no Food and Drug Administration-approved treatments. The ability of drugs that act in the brain in a similar manner to cocaine, but with lower abuse liability, has clinical implications for a treatment of CUD.
Collapse
Affiliation(s)
- Omeed Rahimi
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (O.R., S.R.C., L.J.P., M.A.N.); Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland (J.C., J.L., A.H.N.); Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, Maryland (J.L., R.R.); and EncepHeal Therapeutics, Inc., Winston-Salem, North Carolina (O.R., S.R.C.)
| | - Jianjing Cao
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (O.R., S.R.C., L.J.P., M.A.N.); Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland (J.C., J.L., A.H.N.); Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, Maryland (J.L., R.R.); and EncepHeal Therapeutics, Inc., Winston-Salem, North Carolina (O.R., S.R.C.)
| | - Jenny Lam
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (O.R., S.R.C., L.J.P., M.A.N.); Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland (J.C., J.L., A.H.N.); Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, Maryland (J.L., R.R.); and EncepHeal Therapeutics, Inc., Winston-Salem, North Carolina (O.R., S.R.C.)
| | - Steven R Childers
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (O.R., S.R.C., L.J.P., M.A.N.); Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland (J.C., J.L., A.H.N.); Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, Maryland (J.L., R.R.); and EncepHeal Therapeutics, Inc., Winston-Salem, North Carolina (O.R., S.R.C.)
| | - Rana Rais
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (O.R., S.R.C., L.J.P., M.A.N.); Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland (J.C., J.L., A.H.N.); Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, Maryland (J.L., R.R.); and EncepHeal Therapeutics, Inc., Winston-Salem, North Carolina (O.R., S.R.C.)
| | - Linda J Porrino
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (O.R., S.R.C., L.J.P., M.A.N.); Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland (J.C., J.L., A.H.N.); Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, Maryland (J.L., R.R.); and EncepHeal Therapeutics, Inc., Winston-Salem, North Carolina (O.R., S.R.C.)
| | - Amy Hauck Newman
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (O.R., S.R.C., L.J.P., M.A.N.); Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland (J.C., J.L., A.H.N.); Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, Maryland (J.L., R.R.); and EncepHeal Therapeutics, Inc., Winston-Salem, North Carolina (O.R., S.R.C.)
| | - Michael A Nader
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (O.R., S.R.C., L.J.P., M.A.N.); Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland (J.C., J.L., A.H.N.); Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, Maryland (J.L., R.R.); and EncepHeal Therapeutics, Inc., Winston-Salem, North Carolina (O.R., S.R.C.)
| |
Collapse
|
7
|
Aggarwal S, Mortensen OV. Discovery and Development of Monoamine Transporter Ligands. ADVANCES IN NEUROBIOLOGY 2023; 30:101-129. [PMID: 36928847 PMCID: PMC10074400 DOI: 10.1007/978-3-031-21054-9_4] [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] [Indexed: 03/18/2023]
Abstract
Monoamine transporters (MATs) are targets of a wide range of compounds that have been developed as therapeutic treatments for various neuropsychiatric and neurodegenerative disorders such as depression, ADHD, neuropathic pain, anxiety disorders, stimulant use disorders, epilepsy, and Parkinson's disease. The MAT family is comprised of three main members - the dopamine transporter (DAT), the norepinephrine transporter (NET), and the serotonin transporter (SERT). These transporters are through reuptake responsible for the clearance of their respective monoamine substrates from the extracellular space. The determination of X-ray crystal structures of MATs and their homologues bound with various substrates and ligands has resulted in a surge of structure-function-based studies of MATs to understand the molecular basis of transport function and the mechanism of various ligands that ultimately result in their behavioral effects. This review focusses on recent examples of ligand-based structure-activity relationship studies trying to overcome some of the challenges associated with previously developed MAT inhibitors. These studies have led to the discovery of unique and novel structurally diverse MAT ligands including allosteric modulators. These novel molecular scaffolds serve as leads for designing more effective therapeutic interventions by modulating the activities of MATs and ultimately their associated neurotransmission and behavioral effects.
Collapse
Affiliation(s)
- Shaili Aggarwal
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA.
| | - Ole Valente Mortensen
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Ashraf-Uz-Zaman M, Ji G, Tidwell D, Yin L, Thakolwiboon S, Pan J, Junell R, Griffin Z, Shahi S, Barthels D, Sajib MS, Trippier PC, Mikelis CM, Das H, Avila M, Neugebauer V, German NA. Evaluation of Urea-Based Inhibitors of the Dopamine Transporter Using the Experimental Autoimmune Encephalomyelitis Model of Multiple Sclerosis. ACS Chem Neurosci 2022; 13:217-228. [PMID: 34978174 DOI: 10.1021/acschemneuro.1c00647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The dopaminergic system is involved in the regulation of immune responses in various homeostatic and disease conditions. For conditions such as Parkinson's disease and multiple sclerosis (MS), pharmacological modulation of dopamine (DA) system activity is thought to have therapeutic relevance, providing the basis for using dopaminergic agents as a treatment of relevant states. In particular, it was proposed that restoration of DA levels may inhibit neuroinflammation. We have recently reported a new class of dopamine transporter (DAT) inhibitors with high selectivity to the DAT over other G-protein coupled receptors tested. Here, we continue their evaluation as monoamine transporter inhibitors. Furthermore, we show that the urea-like DAT inhibitor (compound 5) has statistically significant anti-inflammatory effects and attenuates motor deficits and pain behaviors in the experimental autoimmune encephalomyelitis model mimicking clinical signs of MS. To the best of our knowledge, this is the first study reporting the beneficial effects of DAT inhibitor-based treatment in animals with induced autoimmune encephalomyelitis, and the observed results provide additional support to the model of DA-related neuroinflammation.
Collapse
Affiliation(s)
- Md Ashraf-Uz-Zaman
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Guangchen Ji
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Dalton Tidwell
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Linda Yin
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Smathorn Thakolwiboon
- Neurology Department, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Jie Pan
- Neurology Department, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Riley Junell
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Zach Griffin
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Sadisna Shahi
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Derek Barthels
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Md Sanaullah Sajib
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Paul C. Trippier
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Constantinos M. Mikelis
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Hiranmoy Das
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Mirla Avila
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
- Multiple Sclerosis and Demyelinating Diseases Clinic; Department of Neurology, Texas Tech University Health Science Center,Lubbock, Texas 79430, United States
- Neurology Department, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Nadezhda A. German
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| |
Collapse
|
9
|
Reinstatement of synaptic plasticity in the aging brain through specific dopamine transporter inhibition. Mol Psychiatry 2021; 26:7076-7090. [PMID: 34244620 DOI: 10.1038/s41380-021-01214-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aging-related neurological deficits negatively impact mental health, productivity, and social interactions leading to a pronounced socioeconomic burden. Since declining brain dopamine signaling during aging is associated with the onset of neurological impairments, we produced a selective dopamine transporter (DAT) inhibitor to restore endogenous dopamine levels and improve cognitive function. We describe the synthesis and pharmacological profile of (S,S)-CE-158, a highly specific DAT inhibitor, which increases dopamine levels in brain regions associated with cognition. We find both a potentiation of neurotransmission and coincident restoration of dendritic spines in the dorsal hippocampus, indicative of reinstatement of dopamine-induced synaptic plasticity in aging rodents. Treatment with (S,S)-CE-158 significantly improved behavioral flexibility in scopolamine-compromised animals and increased the number of spontaneously active prefrontal cortical neurons, both in young and aging rodents. In addition, (S,S)-CE-158 restored learning and memory recall in aging rats comparable to their young performance in a hippocampus-dependent hole board test. In sum, we present a well-tolerated, highly selective DAT inhibitor that normalizes the age-related decline in cognitive function at a synaptic level through increased dopamine signaling.
Collapse
|
10
|
Gao K, Chen D, Robison AJ, Wei GW. Proteome-Informed Machine Learning Studies of Cocaine Addiction. J Phys Chem Lett 2021; 12:11122-11134. [PMID: 34752088 PMCID: PMC9357290 DOI: 10.1021/acs.jpclett.1c03133] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
No anti-cocaine addiction drugs have been approved by the Food and Drug Administration despite decades of effort. The main challenge is the intricate molecular mechanisms of cocaine addiction, involving synergistic interactions among proteins upstream and downstream of the dopamine transporter. However, it is difficult to study so many proteins with traditional experiments, highlighting the need for innovative strategies in the field. We propose a proteome-informed machine learning (ML) platform for discovering nearly optimal anti-cocaine addiction lead compounds. We analyze proteomic protein-protein interaction networks for cocaine dependence to identify 141 involved drug targets and build 32 ML models for cross-target analysis of more than 60,000 drug candidates or experimental drugs for side effects and repurposing potentials. We further predict their ADMET (absorption, distribution, metabolism, excretion, and toxicity) properties. Our platform reveals that essentially all of the existing drug candidates fail in our cross-target and ADMET screenings but identifies several nearly optimal leads for further optimization.
Collapse
Affiliation(s)
- Kaifu Gao
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Dong Chen
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Alfred J Robison
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
11
|
Van Puyvelde M, Van Cutsem J, Lacroix E, Pattyn N. A State-of-the-Art Review on the Use of Modafinil as A Performance-enhancing Drug in the Context of Military Operationality. Mil Med 2021; 187:52-64. [PMID: 34632515 DOI: 10.1093/milmed/usab398] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/09/2021] [Accepted: 09/20/2021] [Indexed: 11/14/2022] Open
Abstract
INTRODUCTION Modafinil is an eugeroic drug that has been examined to maintain or recover wakefulness, alertness, and cognitive performance when sleep deprived. In a nonmilitary context, the use of modafinil as a nootropic or smart drug, i.e., to improve cognitive performance without being sleep deprived, increases. Although cognitive performance is receiving more explicit attention in a military context, research into the impact of modafinil as a smart drug in function of operationality is lacking. Therefore, the current review aimed at presenting a current state-of-the-art and research agenda on modafinil as a smart drug. Beside the question whether modafinil has an effect or not on cognitive performance, we examined four research questions based on the knowledge on modafinil in sleep-deprived subjects: (1) Is there a difference between the effect of modafinil as a smart drug when administered in repeated doses versus one single dose?; (2) Is the effect of modafinil as a smart drug dose-dependent?; (3) Are there individual-related and/or task-related impact factors?; and (4) What are the reported mental and/or somatic side effects of modafinil as a smart drug? METHOD We conducted a systematic search of the literature in the databases PubMed, Web of Science, and Scopus, using the search terms "Modafinil" and "Cognitive enhance*" in combination with specific terms related to the research questions. The inclusion criteria were studies on healthy human subjects with quantifiable cognitive outcome based on cognitive tasks. RESULTS We found no literature on the impact of a repeated intake of modafinil as a smart drug, although, in users, intake occurs on a regular basis. Moreover, although modafinil was initially said to comprise no risk for abuse, there are now indications that modafinil works on the same neurobiological mechanisms as other addictive stimulants. There is also no thorough research into a potential risk for overconfidence, whereas this risk was identified in sleep-deprived subjects. Furthermore, eventual enhancing effects were beneficial only in persons with an initial lower performance level and/or performing more difficult tasks and modafinil has an adverse effect when used under time pressure and may negatively impact physical performance. Finally, time-on-task may interact with the dose taken. DISCUSSION The use of modafinil as a smart drug should be examined in function of different military profiles considering their individual performance level and the task characteristics in terms of cognitive demands, physical demands, and sleep availability. It is not yet clear to what extent an improvement in one component (e.g., cognitive performance) may negatively affect another component (e.g., physical performance). Moreover, potential risks for abuse and overconfidence in both regular and occasional intake should be thoroughly investigated to depict the trade-off between user benefits and unwanted side effects. We identified that there is a current risk to the field, as this trade-off has been deemed acceptable for sleep-deprived subjects (considering the risk of sleep deprivation to performance) but this reasoning cannot and should not be readily transposed to non-sleep-deprived individuals. We thus conclude against the use of modafinil as a cognitive enhancer in military contexts that do not involve sleep deprivation.
Collapse
Affiliation(s)
- Martine Van Puyvelde
- VIPER Research Unit, Department of LIFE, Royal Military Academy, Brussels 1000, Belgium.,Brain, Body and Cognition, Department of Psychology, Vrije Universiteit Brussel, Brussels 1050, Belgium.,Clinical & Lifespan Psychology, Department of Psychology and Educational Sciences, Vrije Universiteit Brussel, Brussels 1050, Belgium
| | - Jeroen Van Cutsem
- VIPER Research Unit, Department of LIFE, Royal Military Academy, Brussels 1000, Belgium.,MFYS-BLITS, Department of Human Physiology, Vrije Universiteit Brussel, Brussels 1050, Belgium
| | - Emilie Lacroix
- VIPER Research Unit, Department of LIFE, Royal Military Academy, Brussels 1000, Belgium
| | - Nathalie Pattyn
- VIPER Research Unit, Department of LIFE, Royal Military Academy, Brussels 1000, Belgium.,MFYS-BLITS, Department of Human Physiology, Vrije Universiteit Brussel, Brussels 1050, Belgium
| |
Collapse
|
12
|
Lee KH, Fant AD, Guo J, Guan A, Jung J, Kudaibergenova M, Miranda WE, Ku T, Cao J, Wacker S, Duff HJ, Newman AH, Noskov SY, Shi L. Toward Reducing hERG Affinities for DAT Inhibitors with a Combined Machine Learning and Molecular Modeling Approach. J Chem Inf Model 2021; 61:4266-4279. [PMID: 34420294 DOI: 10.1021/acs.jcim.1c00856] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Psychostimulant drugs, such as cocaine, inhibit dopamine reuptake via blockading the dopamine transporter (DAT), which is the primary mechanism underpinning their abuse. Atypical DAT inhibitors are dissimilar to cocaine and can block cocaine- or methamphetamine-induced behaviors, supporting their development as part of a treatment regimen for psychostimulant use disorders. When developing these atypical DAT inhibitors as medications, it is necessary to avoid off-target binding that can produce unwanted side effects or toxicities. In particular, the blockade of a potassium channel, human ether-a-go-go (hERG), can lead to potentially lethal ventricular tachycardia. In this study, we established a counter screening platform for DAT and against hERG binding by combining machine learning-based quantitative structure-activity relationship (QSAR) modeling, experimental validation, and molecular modeling and simulations. Our results show that the available data are adequate to establish robust QSAR models, as validated by chemical synthesis and pharmacological evaluation of a validation set of DAT inhibitors. Furthermore, the QSAR models based on subsets of the data according to experimental approaches used have predictive power as well, which opens the door to target specific functional states of a protein. Complementarily, our molecular modeling and simulations identified the structural elements responsible for a pair of DAT inhibitors having opposite binding affinity trends at DAT and hERG, which can be leveraged for rational optimization of lead atypical DAT inhibitors with desired pharmacological properties.
Collapse
Affiliation(s)
- Kuo Hao Lee
- Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Andrew D Fant
- Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Jiqing Guo
- Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Andy Guan
- Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Joslyn Jung
- Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Mary Kudaibergenova
- Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Williams E Miranda
- Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Therese Ku
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Jianjing Cao
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Soren Wacker
- Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary, Alberta T2N 1N4, Canada.,Achlys Inc., 7-126 Li Ka Shing Center for Health and Innovation, Edmonton, Alberta T6G 2E1, Canada
| | - Henry J Duff
- Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Sergei Y Noskov
- Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, United States
| |
Collapse
|
13
|
Hersey M, Bacon AK, Bailey LG, Coggiano MA, Newman AH, Leggio L, Tanda G. Psychostimulant Use Disorder, an Unmet Therapeutic Goal: Can Modafinil Narrow the Gap? Front Neurosci 2021; 15:656475. [PMID: 34121988 PMCID: PMC8187604 DOI: 10.3389/fnins.2021.656475] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/20/2021] [Indexed: 12/11/2022] Open
Abstract
The number of individuals affected by psychostimulant use disorder (PSUD) has increased rapidly over the last few decades resulting in economic, emotional, and physical burdens on our society. Further compounding this issue is the current lack of clinically approved medications to treat this disorder. The dopamine transporter (DAT) is a common target of psychostimulant actions related to their use and dependence, and the recent availability of atypical DAT inhibitors as a potential therapeutic option has garnered popularity in this research field. Modafinil (MOD), which is approved for clinical use for the treatment of narcolepsy and sleep disorders, blocks DAT just like commonly abused psychostimulants. However, preclinical and clinical studies have shown that it lacks the addictive properties (in both behavioral and neurochemical studies) associated with other abused DAT inhibitors. Clinical availability of MOD has facilitated its off-label use for several psychiatric disorders related to alteration of brain dopamine (DA) systems, including PSUD. In this review, we highlight clinical and preclinical research on MOD and its R-enantiomer, R-MOD, as potential medications for PSUD. Given the complexity of PSUD, we have also reported the effects of MOD on psychostimulant-induced appearance of several symptoms that could intensify the severity of the disease (i.e., sleep disorders and impairment of cognitive functions), besides the potential therapeutic effects of MOD on PSUD.
Collapse
Affiliation(s)
- Melinda Hersey
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Amanda K. Bacon
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Lydia G. Bailey
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Mark A. Coggiano
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Amy H. Newman
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Lorenzo Leggio
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
- Clinical Psychoneuroendo- crinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
- National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, National Institutes of Health, Bethesda, MD, United States
| | - Gianluigi Tanda
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
14
|
Haney M, Rubin E, Denson RK, Foltin RW. Modafinil reduces smoked cocaine self-administration in humans: effects vary as a function of cocaine 'priming' and cost. Drug Alcohol Depend 2021; 221:108554. [PMID: 33610094 PMCID: PMC8026732 DOI: 10.1016/j.drugalcdep.2021.108554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/31/2020] [Accepted: 01/05/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND The absence of an FDA-approved medication for the treatment of cocaine use disorder (CUD) may, in part, reflect the varying conditions present when the decision to use cocaine is made, with one medication unlikely to work under all conditions. The objective of this double-blind, placebo-controlled, human laboratory study was to test the effects of modafinil, a medication with mixed efficacy for the treatment of CUD, using a novel self-administration procedure designed to model distinct clinical scenarios. METHODS During modafinil maintenance (0, 300 mg/day), participants chose to self-administer up to 7 doses of smoked cocaine (25 mg) under 9 conditions: immediately after exposure to: (a) cues associated with cocaine and a non-contingent cocaine administration, i.e. 'prime' (25 mg), (b) only cocaine cues, and (c) neither cues nor cocaine. Each condition was tested when self-administered cocaine cost $5, $10 and $15/dose. RESULTS Nontreatment-seeking cocaine smokers (3 F,13 M), spending $388 ± 218/week on cocaine and with no history of alcohol use disorder, completed the study. Relative to placebo, modafinil robustly attenuated self-administration when cocaine was expensive ($10,$15/dose) and when there was no 'prime.' Modafinil had no effect on self-administration when cocaine was inexpensive ($5/dose) or when participants received a 'prime.' CONCLUSIONS Modafinil's effects on cocaine-taking varied substantially as a function of recent cocaine exposure and cost, which may help explain the mixed clinical findings. Modafinil may be most effective for preventing relapse in abstinent patients, particularly under conditions in which cocaine is costly, rather than initiating abstinence for those continuing to use cocaine.
Collapse
Affiliation(s)
- Margaret Haney
- Department of Psychiatry, Columbia University Irving Medical Center and the New York State Psychiatric Institute, USA.
| | - Eric Rubin
- Department of Psychiatry, Harlem Hospital Center, Columbia University College of Physicians and Surgeons, USA
| | - Rebecca K Denson
- Department of Psychiatry, Columbia University Irving Medical Center and the New York State Psychiatric Institute, USA
| | - Richard W Foltin
- Department of Psychiatry, Columbia University Irving Medical Center and the New York State Psychiatric Institute, USA
| |
Collapse
|
15
|
Newman AH, Ku T, Jordan CJ, Bonifazi A, Xi ZX. New Drugs, Old Targets: Tweaking the Dopamine System to Treat Psychostimulant Use Disorders. Annu Rev Pharmacol Toxicol 2021; 61:609-628. [PMID: 33411583 PMCID: PMC9341034 DOI: 10.1146/annurev-pharmtox-030220-124205] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The abuse of illicit psychostimulants such as cocaine and methamphetamine continues to pose significant health and societal challenges. Despite considerable efforts to develop medications to treat psychostimulant use disorders, none have proven effective, leaving an underserved patient population and unanswered questions about what mechanism(s) of action should be targeted for developing pharmacotherapies. As both cocaine and methamphetamine rapidly increase dopamine (DA) levels in mesolimbic brain regions, leading to euphoria that in some can lead to addiction, targets in which this increased dopaminergic tone may be mitigated have been explored. Further, understanding and targeting mechanisms underlying relapse are fundamental to the success of discovering medications that reduce the reinforcing effects of the drug of abuse, decrease the negative reinforcement or withdrawal/negative affect that occurs during abstinence, or both. Atypical inhibitors of the DA transporter and partial agonists/antagonists at DA D3 receptors are described as two promising targets for future drug development.
Collapse
Affiliation(s)
- Amy Hauck Newman
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA;
| | - Therese Ku
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA;
| | - Chloe J Jordan
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA;
| | - Alessandro Bonifazi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA;
| | - Zheng-Xiong Xi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA;
| |
Collapse
|
16
|
Improving translation of animal models of addiction and relapse by reverse translation. Nat Rev Neurosci 2020; 21:625-643. [PMID: 33024318 DOI: 10.1038/s41583-020-0378-z] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2020] [Indexed: 12/13/2022]
Abstract
Critical features of human addiction are increasingly being incorporated into complementary animal models, including escalation of drug intake, punished drug seeking and taking, intermittent drug access, choice between drug and non-drug rewards, and assessment of individual differences based on criteria in the fourth edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM-IV). Combined with new technologies, these models advanced our understanding of brain mechanisms of drug self-administration and relapse, but these mechanistic gains have not led to improvements in addiction treatment. This problem is not unique to addiction neuroscience, but it is an increasing source of disappointment and calls to regroup. Here we first summarize behavioural and neurobiological results from the animal models mentioned above. We then propose a reverse translational approach, whose goal is to develop models that mimic successful treatments: opioid agonist maintenance, contingency management and the community-reinforcement approach. These reverse-translated 'treatments' may provide an ecologically relevant platform from which to discover new circuits, test new medications and improve translation.
Collapse
|
17
|
Rotolo RA, Presby RE, Tracy O, Asar S, Yang JH, Correa M, Murray F, Salamone JD. The novel atypical dopamine transport inhibitor CT-005404 has pro-motivational effects in neurochemical and inflammatory models of effort-based dysfunctions related to psychopathology. Neuropharmacology 2020; 183:108325. [PMID: 32956676 DOI: 10.1016/j.neuropharm.2020.108325] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/11/2020] [Accepted: 09/15/2020] [Indexed: 01/23/2023]
Abstract
Depressed individuals suffer from effort-related motivational symptoms such as anergia and fatigue, which are resistant to treatment with many common antidepressants. While drugs that block dopamine transport (DAT) reportedly have positive motivational effects, DAT inhibitors such as cocaine and amphetamines produce undesirable side effects. Thus, there is a need to develop and characterize novel atypical DAT inhibitors with unique and selective binding profiles. Rodent effort-based choice tasks provide useful models of motivational dysfunctions. With these tasks, animals choose between a high-effort instrumental action leading to highly valued reinforcement vs. a low effort/low reward option. The present studies focused on the initial characterization of a novel atypical DAT inhibitor, CT-005404, which binds to DAT with high selectivity relative to serotonin and norepinephrine transport, and produces long-term elevations of extracellular DA. CT-005404 was assessed for its ability to attenuate the effort-related motivational effects of the DA depleting agent tetrabenazine and the pro-inflammatory cytokine interleukin-1β (IL-1β) using a fixed ratio 5/chow feeding choice test. Tetrabenazine (1.0 mg/kg i.p.) shifted choice behavior, decreasing lever pressing and increasing chow intake. IL-1β (4.0 μg/kg i.p.) also decreased lever pressing. CT-005404 was co-administered (7.5-30.0 mg/kg p.o.) with either tetrabenazine or IL-1β, and the 15.0 and 30.0 mg/kg doses significantly reversed the effects of tetrabenazine and IL-1β. CT-005404 administered alone produced a dose-related increase in lever pressing in rats tested on a progressive ratio/chow feeding choice task. Atypical DAT inhibitors such as CT-005404 offer potential as a new avenue for drug treatment of motivational dysfunctions in humans.
Collapse
Affiliation(s)
- Renee A Rotolo
- Department of Psychological Sciences, University of Connecticut, Storrs, CT, 06261-1020, USA
| | - Rose E Presby
- Department of Psychological Sciences, University of Connecticut, Storrs, CT, 06261-1020, USA
| | - Olivia Tracy
- Department of Psychological Sciences, University of Connecticut, Storrs, CT, 06261-1020, USA
| | - Sokaina Asar
- Department of Psychological Sciences, University of Connecticut, Storrs, CT, 06261-1020, USA
| | - Jen-Hau Yang
- Department of Psychological Sciences, University of Connecticut, Storrs, CT, 06261-1020, USA
| | - Merce Correa
- Department of Psychological Sciences, University of Connecticut, Storrs, CT, 06261-1020, USA; Àrea de Psicobiologia, Campus de Riu Sec, Universitat Jaume I, 12071, Castelló, Spain
| | - Fraser Murray
- Chronos Therapeutics, The Magdalen Centre, Oxford Science Park, Oxford, OX4 4GA, UK
| | - John D Salamone
- Department of Psychological Sciences, University of Connecticut, Storrs, CT, 06261-1020, USA.
| |
Collapse
|
18
|
Tanda G, Hersey M, Hempel B, Xi ZX, Newman AH. Modafinil and its structural analogs as atypical dopamine uptake inhibitors and potential medications for psychostimulant use disorder. Curr Opin Pharmacol 2020; 56:13-21. [PMID: 32927246 DOI: 10.1016/j.coph.2020.07.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 12/20/2022]
Abstract
Pharmacotherapeutics for treatment of psychostimulant use disorder are still an unmet medical goal. Recently, off label use of modafinil (MOD), an approved medication for treatment of sleep disturbances, has been tested as a therapeutic for cocaine and methamphetamine use disorder. Positive results have been found in subjects dependent on psychostimulants without concurrent abuse of other substances. Novel structural analogs of MOD have been synthesized in the search for compounds with potentially broader therapeutic efficacy than the parent drug. In the present report we review their potential efficacy as treatments for psychostimulant abuse and dependence assessed in preclinical tests. Results from these preclinical proof of concept studies reveal that some modafinil analogs do not possess typical cocaine-like neurochemical and behavioral effects. Further, they might blunt the reinforcing effects of psychostimulants in animal models, suggesting their potential efficacy as pharmacotherapeutics for treatment of psychostimulant use disorders.
Collapse
Affiliation(s)
- Gianluigi Tanda
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, NIH, DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA.
| | - Melinda Hersey
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, NIH, DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Briana Hempel
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, NIH, DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Zheng-Xiong Xi
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, NIH, DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Amy Hauck Newman
- Medication Development Program, Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, NIH, DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA
| |
Collapse
|
19
|
Roberts CA, Jones A, Sumnall H, Gage SH, Montgomery C. How effective are pharmaceuticals for cognitive enhancement in healthy adults? A series of meta-analyses of cognitive performance during acute administration of modafinil, methylphenidate and D-amphetamine. Eur Neuropsychopharmacol 2020; 38:40-62. [PMID: 32709551 DOI: 10.1016/j.euroneuro.2020.07.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/11/2020] [Accepted: 07/06/2020] [Indexed: 12/20/2022]
Abstract
Modafinil, methyphenidate (MPH) and d-amphetamine (d-amph) are putative cognitive enhancers. However, efficacy of cognitive enhancement has yet to be fully established. We examined cognitive performance in healthy non-sleep-deprived adults following modafinil, MPH, or d-amph vs placebo in 3 meta-analyses, using subgroup analysis by cognitive domain; executive functions (updating, switching, inhibitory control, access to semantic/long term memory), spatial working memory, recall, selective attention, and sustained attention. We adhered to PRISMA. We identified k = 47 studies for analysis; k = 14 studies (64 effect sizes) for modafinil, k = 24 studies (47 effect sizes) for Methylphenidate, and k = 10 (27 effect sizes) for d-amph. There was an overall effect of modafinil (SMD=0.12, p=.01). Modafinil improved memory updating (SMD=0.28, p=.03). There was an overall effect of MPH (SMD=0.21, p=.0004) driven by improvements in recall (SMD=0.43, p=.0002), sustained attention (SMD=0.42, p=.0004), and inhibitory control (SMD=0.27, p=.03). There were no effects for d-amph. MPH and modafinil show enhancing effects in specific sub-domains of cognition. However, data with these stimulants is far from positive if we consider that effects are small, in experiments that do not accurately reflect their actual use in the wider population. There is a user perception that these drugs are effective cognitive enhancers, but this is not supported by the evidence so far.
Collapse
Affiliation(s)
- Carl A Roberts
- Department of Psychological Sciences, University of Liverpool, United Kingdom.
| | - Andrew Jones
- Department of Psychological Sciences, University of Liverpool, United Kingdom
| | - Harry Sumnall
- Public Health Institute, Liverpool John Moores University, Liverpool, United Kingdom
| | - Suzanne H Gage
- Department of Psychological Sciences, University of Liverpool, United Kingdom
| | - Catharine Montgomery
- Department of Psychology, John Moores Liverpool University, Liverpool, United Kingdom
| |
Collapse
|
20
|
Sousa A, Dinis-Oliveira RJ. Pharmacokinetic and pharmacodynamic of the cognitive enhancer modafinil: Relevant clinical and forensic aspects. Subst Abus 2020; 41:155-173. [PMID: 31951804 DOI: 10.1080/08897077.2019.1700584] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Modafinil is a nonamphetamine nootropic drug with an increasingly therapeutic interest due to its different sites of action and behavioral effects in comparison to cocaine or amphetamine. A review of modafinil (and of its prodrug adrafinil and its R-enantiomer armodafinil) chemical, pharmacokinetic, pharmacodynamic, toxicological, clinical and forensic aspects was performed, aiming to better understand possible health problems associated to its unconscious and unruled use. Modafinil is a racemate metabolized mainly in the liver into its inactive acid and sulfone metabolites, which undergo primarily renal excretion. Although not fully clarified, major effects seem to be associated to inhibition of dopamine reuptake and modulation of several other neurochemical pathways, namely noradrenergic, serotoninergic, orexinergic, histaminergic, glutamatergic and GABAergic. Due its wake-promoting effects, modafinil is used for the treatment of daily sleepiness associated to narcolepsy, obstructive sleep apnea and shift work sleep disorder. Its psychotropic and cognitive effects are also attractive in several other pathologies and conditions that affect sleep structure, induce fatigue and lethargy, and impair cognitive abilities. Additionally, in health subjects, including students, modafinil is being used off-label to overcome sleepiness, increase concentration and improve cognitive potential. The most common adverse effects associated to modafinil intake are headache, insomnia, anxiety, diarrhea, dry mouth and raise in blood pressure and heart rate. Infrequently, severe dermatologic effects in children, including maculopapular and morbilliform rash, erythema multiforme and Stevens-Johnson Syndrome have been reported. Intoxication and dependence associated to modafinil are uncommon. Further research on effects and health implications of modafinil and its analogs is steel needed to create evidence-based policies.
Collapse
Affiliation(s)
- Ana Sousa
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Ricardo Jorge Dinis-Oliveira
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, Porto, Portugal.,IINFACTS - Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, Gandra, Portugal.,UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
21
|
Kalaba P, Ilić M, Aher NY, Dragačević V, Wieder M, Zehl M, Wackerlig J, Beyl S, Sartori SB, Ebner K, Roller A, Lukic N, Beryozkina T, Gonzalez ERP, Neill P, Khan JA, Bakulev V, Leban JJ, Hering S, Pifl C, Singewald N, Lubec J, Urban E, Sitte HH, Langer T, Lubec G. Structure-Activity Relationships of Novel Thiazole-Based Modafinil Analogues Acting at Monoamine Transporters. J Med Chem 2019; 63:391-417. [PMID: 31841637 DOI: 10.1021/acs.jmedchem.9b01938] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Atypical dopamine reuptake inhibitors, such as modafinil, are used for the treatment of sleeping disorders and investigated as potential therapeutics against cocaine addiction and for cognitive enhancement. Our continuous effort to find modafinil analogues with higher inhibitory activity on and selectivity toward the dopamine transporter (DAT) has previously led to the promising thiazole-containing derivatives CE-103, CE-111, CE-123, and CE-125. Here, we describe the synthesis and activity of a series of compounds based on these scaffolds, which resulted in several new selective DAT inhibitors and gave valuable insights into the structure-activity relationships. Introduction of the second chiral center and subsequent chiral separations provided all four stereoisomers, whereby the S-configuration on both generally exerted the highest activity and selectivity on DAT. The representative compound of this series was further characterized by in silico, in vitro, and in vivo studies that have demonstrated both safety and efficacy profile of this compound class.
Collapse
Affiliation(s)
- Predrag Kalaba
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences , University of Vienna , Althanstraße 14 , 1090 Vienna , Austria
| | - Marija Ilić
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences , University of Vienna , Althanstraße 14 , 1090 Vienna , Austria
| | - Nilima Y Aher
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences , University of Vienna , Althanstraße 14 , 1090 Vienna , Austria
| | - Vladimir Dragačević
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences , University of Vienna , Althanstraße 14 , 1090 Vienna , Austria
| | - Marcus Wieder
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences , University of Vienna , Althanstraße 14 , 1090 Vienna , Austria
| | - Martin Zehl
- Department of Analytical Chemistry, Faculty of Chemistry , University of Vienna , Währinger Straße 38 , 1090 Vienna , Austria
| | - Judith Wackerlig
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences , University of Vienna , Althanstraße 14 , 1090 Vienna , Austria
| | - Stanislav Beyl
- Department of Pharmacology and Toxicology, Faculty of Life Sciences , University of Vienna , Althanstraße 14 , 1090 Vienna , Austria
| | - Simone B Sartori
- Center for Molecular Biosciences Innsbruck (CMBI), Department of Pharmacology and Toxicology, Institute of Pharmacy , Leopold Franzens University Innsbruck , 6020 Innsbruck , Austria
| | - Karl Ebner
- Center for Molecular Biosciences Innsbruck (CMBI), Department of Pharmacology and Toxicology, Institute of Pharmacy , Leopold Franzens University Innsbruck , 6020 Innsbruck , Austria
| | - Alexander Roller
- X-ray Structure Analysis Centre, Faculty of Chemistry , University of Vienna , Währinger Straße 38 , 1090 Vienna , Austria
| | - Natalie Lukic
- X-ray Structure Analysis Centre, Faculty of Chemistry , University of Vienna , Währinger Straße 38 , 1090 Vienna , Austria
| | - Tetyana Beryozkina
- Ural Federal University Named after the First President of Russia B. N. Yeltsin , 19 Mira st. , Yekaterinburg 620002 , Russia
| | - Eduardo Rene Perez Gonzalez
- Laboratory of Fine Organic Chemistry, Department of Chemistry and Biochemistry, Faculty of Science and Technology , University of Sao Paulo State , Roberto Simonsen 305 , CEP 19060-900 , Presidente Prudente , SP , Brazil
| | - Philip Neill
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences , University of Vienna , Althanstraße 14 , 1090 Vienna , Austria
| | - Jawad Akbar Khan
- Institute of Pharmacology, Centre of Physiology and Pharmacology , Medical University of Vienna , 1090 Vienna , Austria
| | - Vasiliy Bakulev
- Ural Federal University Named after the First President of Russia B. N. Yeltsin , 19 Mira st. , Yekaterinburg 620002 , Russia
| | - Johann Jakob Leban
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences , University of Vienna , Althanstraße 14 , 1090 Vienna , Austria
| | - Steffen Hering
- Department of Pharmacology and Toxicology, Faculty of Life Sciences , University of Vienna , Althanstraße 14 , 1090 Vienna , Austria
| | - Christian Pifl
- Centre for Brain Research , Medical University of Vienna , 1090 Vienna , Austria
| | - Nicolas Singewald
- Center for Molecular Biosciences Innsbruck (CMBI), Department of Pharmacology and Toxicology, Institute of Pharmacy , Leopold Franzens University Innsbruck , 6020 Innsbruck , Austria
| | - Jana Lubec
- Neuroscience Laboratory , Paracelsus Medical University , A-5020 Salzburg , Austria
| | - Ernst Urban
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences , University of Vienna , Althanstraße 14 , 1090 Vienna , Austria
| | - Harald H Sitte
- Institute of Pharmacology, Centre of Physiology and Pharmacology , Medical University of Vienna , 1090 Vienna , Austria
| | - Thierry Langer
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences , University of Vienna , Althanstraße 14 , 1090 Vienna , Austria
| | - Gert Lubec
- Neuroscience Laboratory , Paracelsus Medical University , A-5020 Salzburg , Austria
| |
Collapse
|
22
|
Jordan CJ, Cao J, Newman AH, Xi ZX. Progress in agonist therapy for substance use disorders: Lessons learned from methadone and buprenorphine. Neuropharmacology 2019; 158:107609. [PMID: 31009632 PMCID: PMC6745247 DOI: 10.1016/j.neuropharm.2019.04.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/25/2019] [Accepted: 04/12/2019] [Indexed: 12/12/2022]
Abstract
Substance use disorders (SUD) are serious public health problems worldwide. Although significant progress has been made in understanding the neurobiology of drug reward and the transition to addiction, effective pharmacotherapies for SUD remain limited and a majority of drug users relapse even after a period of treatment. The United States Food and Drug Administration (FDA) has approved several medications for opioid, nicotine, and alcohol use disorders, whereas none are approved for the treatment of cocaine or other psychostimulant use disorders. The medications approved by the FDA for the treatment of SUD can be divided into two major classes - agonist replacement therapies, such as methadone and buprenorphine for opioid use disorders (OUD), nicotine replacement therapy (NRT) and varenicline for nicotine use disorders (NUD), and antagonist therapies, such as naloxone for opioid overdose and naltrexone for promoting abstinence. In the present review, we primarily focus on the pharmacological rationale of agonist replacement strategies in treatment of opioid dependence, and the potential translation of this rationale to new therapies for cocaine use disorders. We begin by describing the neural mechanisms underlying opioid reward, followed by preclinical and clinical findings supporting the utility of agonist therapies in the treatment of OUD. We then discuss recent progress of agonist therapies for cocaine use disorders based on lessons learned from methadone and buprenorphine. We contend that future studies should identify agonist pharmacotherapies that can facilitate abstinence in patients who are motivated to quit their illicit drug use. Focusing on those that are able to achieve abstinence from cocaine will provide a platform to broaden the effectiveness of medication and psychosocial treatment strategies for this underserved population. This article is part of the Special Issue entitled 'New Vistas in Opioid Pharmacology'.
Collapse
Affiliation(s)
- Chloe J Jordan
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Jianjing Cao
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Amy Hauck Newman
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Zheng-Xiong Xi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA.
| |
Collapse
|
23
|
Newman AH, Cao J, Keighron JD, Jordan CJ, Bi GH, Liang Y, Abramyan AM, Avelar AJ, Tschumi CW, Beckstead MJ, Shi L, Tanda G, Xi ZX. Translating the atypical dopamine uptake inhibitor hypothesis toward therapeutics for treatment of psychostimulant use disorders. Neuropsychopharmacology 2019; 44:1435-1444. [PMID: 30858517 PMCID: PMC6785152 DOI: 10.1038/s41386-019-0366-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/27/2019] [Accepted: 02/27/2019] [Indexed: 11/10/2022]
Abstract
Medication-assisted treatments are unavailable to patients with cocaine use disorders. Efforts to develop potential pharmacotherapies have led to the identification of a promising lead molecule, JJC8-091, that demonstrates a novel binding mode at the dopamine transporter (DAT). Here, JJC8-091 and a structural analogue, JJC8-088, were extensively and comparatively assessed to elucidate neurochemical correlates to their divergent behavioral profiles. Despite sharing significant structural similarity, JJC8-088 was more cocaine-like, increasing extracellular DA concentrations in the nucleus accumbens shell (NAS) efficaciously and more potently than JJC8-091. In contrast, JJC8-091 was not self-administered and was effective in blocking cocaine-induced reinstatement to drug seeking. Electrophysiology experiments confirmed that JJC8-091 was more effective than JJC8-088 at inhibiting cocaine-mediated enhancement of DA neurotransmission. Further, when VTA DA neurons in DAT-cre mice were optically stimulated, JJC8-088 produced a significant leftward shift in the stimulation-response curve, similar to cocaine, while JJC8-091 shifted the curve downward, suggesting attenuation of DA-mediated brain reward. Computational models predicted that JJC8-088 binds in an outward facing conformation of DAT, similar to cocaine. Conversely, JJC8-091 steers DAT towards a more occluded conformation. Collectively, these data reveal the underlying molecular mechanism at DAT that may be leveraged to rationally optimize leads for the treatment of cocaine use disorders, with JJC8-091 representing a compelling candidate for development.
Collapse
Affiliation(s)
- Amy Hauck Newman
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD, 21224, USA.
| | - Jianjing Cao
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Jacqueline D. Keighron
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Chloe J. Jordan
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Guo-Hua Bi
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Ying Liang
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Ara M. Abramyan
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Alicia J. Avelar
- 0000 0001 0629 5880grid.267309.9Department of Cellular and Integrative Physiology, UT Health Science Center, San Antonio, TX USA
| | - Christopher W. Tschumi
- 0000 0001 0629 5880grid.267309.9Department of Cellular and Integrative Physiology, UT Health Science Center, San Antonio, TX USA ,0000 0000 8527 6890grid.274264.1Aging & Metabolism Research Group, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Michael J. Beckstead
- 0000 0001 0629 5880grid.267309.9Department of Cellular and Integrative Physiology, UT Health Science Center, San Antonio, TX USA ,0000 0000 8527 6890grid.274264.1Aging & Metabolism Research Group, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Lei Shi
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Gianluigi Tanda
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Zheng-Xiong Xi
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| |
Collapse
|
24
|
Keighron JD, Quarterman JC, Cao J, DeMarco EM, Coggiano MA, Gleaves A, Slack RD, Zanettini C, Newman AH, Tanda G. Effects of ( R)-Modafinil and Modafinil Analogues on Dopamine Dynamics Assessed by Voltammetry and Microdialysis in the Mouse Nucleus Accumbens Shell. ACS Chem Neurosci 2019; 10:2012-2021. [PMID: 30645944 DOI: 10.1021/acschemneuro.8b00340] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Recent discoveries have improved our understanding of the physiological and pathological roles of the dopamine transporter (DAT); however, only a few drugs are clinically available for DAT-implicated disorders. Among those drugs, modafinil (MOD) and its ( R)-enantiomer (R-MOD) have been used off-label as therapies for psychostimulant use disorders, but they have shown limited effectiveness in clinical trials. Recent preclinical studies on MOD and R-MOD have led to chemically modified structures aimed toward improving their neurobiological properties that might lead to more effective therapeutics for stimulant use disorders. This study examines three MOD analogues (JJC8-016, JJC8-088, and JJC8-091) with improved DAT affinities compared to their parent compound. These compounds were investigated for their effects on the neurochemistry (brain microdialysis and FSCV) and behavior (ambulatory activity) of male Swiss-Webster mice. Our data indicate that these compounds have dissimilar effects on tonic and phasic dopamine in the nucleus accumbens shell and variability in producing ambulatory activity. These results suggest that small changes in the chemical structure of a DAT inhibitor can cause compounds such as JJC8-088 to produce effects similar to abused psychostimulants like cocaine. In contrast, other compounds like JJC8-091 do not share cocaine-like effects and have a more atypical DAT-inhibitor profile, which may prove to be an advancement in the treatment of psychostimulant use disorders.
Collapse
|
25
|
Tomlinson MJ, Krout D, Pramod AB, Lever JR, Newman AH, Henry LK, Vaughan RA. Identification of the benztropine analog [ 125I]GA II 34 binding site on the human dopamine transporter. Neurochem Int 2018; 123:34-45. [PMID: 30125594 DOI: 10.1016/j.neuint.2018.08.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/10/2018] [Accepted: 08/14/2018] [Indexed: 12/21/2022]
Abstract
The dopamine transporter (DAT) is a neuronal membrane protein that is responsible for reuptake of dopamine (DA) from the synapse and functions as a major determinant in control of DA neurotransmission. Cocaine and many psychostimulant drugs bind to DAT and block reuptake, inducing DA overflow that forms the neurochemical basis for euphoria and addiction. Paradoxically, however, some ligands such as benztropine (BZT) bind to DAT and inhibit reuptake but do not produce these effects, and it has been hypothesized that differential mechanisms of binding may stabilize specific transporter conformations that affect downstream neurochemical or behavioral outcomes. To investigate the binding mechanisms of BZT on DAT we used the photoaffinity BZT analog [125I]N-[n-butyl-4-(4‴-azido-3‴-iodophenyl)]-4',4″-difluoro-3α-(diphenylmethoxy)tropane ([125I]GA II 34) to identify the site of cross-linking and predict the binding pose relative to that of previously-examined cocaine photoaffinity analogs. Biochemical findings show that adduction of [125I]GA II 34 occurs at residues Asp79 or Leu80 in TM1, with molecular modeling supporting adduction to Leu80 and a pharmacophore pose in the central S1 site similar to that of cocaine and cocaine analogs. Substituted cysteine accessibility method protection analyses verified these findings, but identified some differences in structural stabilization relative to cocaine that may relate to BZT neurochemical outcomes.
Collapse
Affiliation(s)
- Michael J Tomlinson
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, United States
| | - Danielle Krout
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, United States
| | - Akula Bala Pramod
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, United States
| | - John R Lever
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, 65201, United States; Department of Radiology and Radiopharmaceutical Sciences Institute, University of Missouri, Columbia, MO 65211, United States
| | - Amy Hauck Newman
- Medicinal Chemistry Section, National Institute on Drug Abuse - Intramural Research Program, Baltimore, MD 21224, United States
| | - L Keith Henry
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, United States.
| | - Roxanne A Vaughan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, United States.
| |
Collapse
|
26
|
Kristofova M, Aher YD, Ilic M, Radoman B, Kalaba P, Dragacevic V, Aher NY, Leban J, Korz V, Zanon L, Neuhaus W, Wieder M, Langer T, Urban E, Sitte HH, Hoeger H, Lubec G, Aradska J. A daily single dose of a novel modafinil analogue CE-123 improves memory acquisition and memory retrieval. Behav Brain Res 2018; 343:83-94. [PMID: 29410048 DOI: 10.1016/j.bbr.2018.01.032] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/28/2018] [Accepted: 01/29/2018] [Indexed: 01/25/2023]
Abstract
Dopamine reuptake inhibitors have been shown to improve cognitive parameters in various tasks and animal models. We recently reported a series of modafinil analogues, of which the most promising, 5-((benzhydrylsulfinyl)methyl) thiazole (CE-123), was selected for further development. The present study aims to characterize pharmacological properties of CE-123 and to investigate the potential to enhance memory performance in a rat model. In vitro transporter assays were performed in cells expressing human transporters. CE-123 blocked uptake of [3H] dopamine (IC50 = 4.606 μM) while effects on serotonin (SERT) and the norepinephrine transporter (NET) were negligible. Blood-brain barrier and pharmacokinetic studies showed that the compound reached the brain and lower elimination than R-modafinil. The Pro-cognitive effect was evaluated in a spatial hole-board task in male Sprague-Dawley rats and CE-123 enhances memory acquisition and memory retrieval, represented by significantly increased reference memory indices and shortened latency. Since DAT blockers can be considered as indirect dopamine receptor agonists, western blotting was used to quantify protein levels of dopamine receptors D1R, D2R and D5R and DAT in the synaptosomal fraction of hippocampal subregions CA1, CA3 and dentate gyrus (DG). CE-123 administration in rats increased total DAT levels and D1R protein levels were significantly increased in CA1 and CA3 in treated/trained groups. The increase of D5R was observed in DG only. Dopamine receptors, particularly D1R, seem to play a role in mediating CE-123-induced memory enhancement. Dopamine reuptake inhibition by CE-123 may represent a novel and improved stimulant therapeutic for impairments of cognitive functions.
Collapse
Affiliation(s)
- Martina Kristofova
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Yogesh D Aher
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Marija Ilic
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Bojana Radoman
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Predrag Kalaba
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Vladimir Dragacevic
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Nilima Y Aher
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Johann Leban
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Volker Korz
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Lisa Zanon
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Winfried Neuhaus
- Competence Unit Molecular Diagnostics, Competence Center Health and Bioresources, Austrian Institute of Technology (AIT) GmbH, Vienna, Austria
| | - Marcus Wieder
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Thierry Langer
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Ernst Urban
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Harald H Sitte
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Harald Hoeger
- Core Unit of Biomedical Research, Division of Laboratory Animal Science and Genetics, Medical University of Vienna, Himberg, Austria
| | - Gert Lubec
- Department of Neuroproteomics, Paracelsus Medical University, Salzburg, Austria.
| | - Jana Aradska
- Department of Neuroproteomics, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
27
|
Abstract
The dopamine (DAT), serotonin (SERT), and norepinephrine (NET) transporters, which are collectively referred to as monoamine transporters (MATs), play significant roles in regulating the neuronal response to these neurotransmitters. MATs terminate the action of these neurotransmitters by translocating them from the synaptic space into the presynaptic neurons. These three transmitters are responsible for controlling a number of physiological, emotional, and behavioral functions, with their transporters being the site of action of drugs employed for the treatment of a variety of conditions, including depression, anxiety, ADHD, schizophrenia, and psychostimulant abuse. Provided in this unit is information on the localization and regulation of MATs and the structural components of these proteins most responsible for the translocation process. Also included is a brief description of the evolution of ligands that interact with these transporters, as well as current theories concerning the pharmacological effects of substances that interact with these sites, including the molecular mechanisms of action of uptake inhibitors and allosteric modulators. Data relating to the presence, structure, and functions of allosteric modulators are included as well. The aim of this review is to provide background information on MATs to those who are new to this field, with a focus on the therapeutic potential of compounds that interact with these substrate transport sites. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Shaili Aggarwal
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Pennsylvania
| | - Ole V Mortensen
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Pennsylvania
| |
Collapse
|
28
|
Tunstall BJ, Ho CP, Cao J, Vendruscolo JCM, Schmeichel BE, Slack RD, Tanda G, Gadiano AJ, Rais R, Slusher BS, Koob GF, Newman AH, Vendruscolo LF. Atypical dopamine transporter inhibitors attenuate compulsive-like methamphetamine self-administration in rats. Neuropharmacology 2017; 131:96-103. [PMID: 29217282 DOI: 10.1016/j.neuropharm.2017.12.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 12/01/2017] [Accepted: 12/03/2017] [Indexed: 11/30/2022]
Abstract
Methamphetamine (METH) is a highly addictive drug, but no pharmacological treatment is yet available for METH use disorders. Similar to METH, the wake-promoting drug (R)-modafinil (R-MOD) binds to the dopamine transporter (DAT). Unlike METH, R-MOD is not a substrate for transport by DAT and has low abuse potential. We tested the hypothesis that the atypical DAT inhibitor R-MOD and compounds that are derived from modafinil would decrease METH intake by reducing the actions of METH at the DAT. We tested the effects of systemic injections of R-MOD and four novel modafinil-derived ligands with increased DAT affinity (JJC8-016, JJC8-088, JJC8-089, and JJC8-091) on intravenous (i.v.) METH self-administration in rats that were allowed short access (ShA; 1 h) or long access (LgA; 6 h) to the drug. ShA rats exhibited stable METH intake over sessions, whereas LgA rats exhibited an escalation of drug intake. R-MOD decreased METH self-administration in ShA and LgA rats (in the 1st hour only). JJC8-091 and JJC8-016 decreased METH self-administration in both ShA and LgA rats. JJC8-089 decreased METH self-administration in LgA rats only, whereas JJC8-088 had no effect on METH self-administration in either ShA or LgA rats. These findings support the potential of atypical DAT inhibitors for the treatment of METH use disorders and suggest several novel compounds as candidate drugs.
Collapse
Affiliation(s)
- Brendan J Tunstall
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Chelsea P Ho
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Jianjing Cao
- Molecular Targets and Medications Discovery Program, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Janaína C M Vendruscolo
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Brooke E Schmeichel
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Rachel D Slack
- Molecular Targets and Medications Discovery Program, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Gianluigi Tanda
- Molecular Targets and Medications Discovery Program, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Alexandra J Gadiano
- Molecular Targets and Medications Discovery Program, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA; Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rana Rais
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Barbara S Slusher
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - George F Koob
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Amy H Newman
- Molecular Targets and Medications Discovery Program, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA.
| | - Leandro F Vendruscolo
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
29
|
Krout D, Pramod AB, Dahal RA, Tomlinson MJ, Sharma B, Foster JD, Zou MF, Boatang C, Newman AH, Lever JR, Vaughan RA, Henry LK. Inhibitor mechanisms in the S1 binding site of the dopamine transporter defined by multi-site molecular tethering of photoactive cocaine analogs. Biochem Pharmacol 2017; 142:204-215. [PMID: 28734777 DOI: 10.1016/j.bcp.2017.07.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/17/2017] [Indexed: 10/19/2022]
Abstract
Dopamine transporter (DAT) blockers like cocaine and many other abused and therapeutic drugs bind and stabilize an inactive form of the transporter inhibiting reuptake of extracellular dopamine (DA). The resulting increases in DA lead to the ability of these drugs to induce psychomotor alterations and addiction, but paradoxical findings in animal models indicate that not all DAT antagonists induce cocaine-like behavioral outcomes. How this occurs is not known, but one possibility is that uptake inhibitors may bind at multiple locations or in different poses to stabilize distinct conformational transporter states associated with differential neurochemical endpoints. Understanding the molecular mechanisms governing the pharmacological inhibition of DAT is therefore key for understanding the requisite interactions for behavioral modulation and addiction. Previously, we leveraged complementary computational docking, mutagenesis, peptide mapping, and substituted cysteine accessibility strategies to identify the specific adduction site and binding pose for the crosslinkable, photoactive cocaine analog, RTI 82, which contains a photoactive azide attached at the 2β position of the tropane pharmacophore. Here, we utilize similar methodology with a different cocaine analog N-[4-(4-azido-3-I-iodophenyl)-butyl]-2-carbomethoxy-3-(4-chlorophenyl)tropane, MFZ 2-24, where the photoactive azide is attached to the tropane nitrogen. In contrast to RTI 82, which crosslinked into residue Phe319 of transmembrane domain (TM) 6, our findings show that MFZ 2-24 adducts to Leu80 in TM1 with modeling and biochemical data indicating that MFZ 2-24, like RTI 82, occupies the central S1 binding pocket with the (+)-charged tropane ring nitrogen coordinating with the (-)-charged carboxyl side chain of Asp79. The superimposition of the tropane ring in the three-dimensional binding poses of these two distinct ligands provides strong experimental evidence for cocaine binding to DAT in the S1 site and the importance of the tropane moiety in competitive mechanisms of DA uptake inhibition. These findings set a structure-function baseline for comparison of typical and atypical DAT inhibitors and how their interactions with DAT could lead to the loss of cocaine-like behaviors.
Collapse
Affiliation(s)
- Danielle Krout
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, USA
| | - Akula Bala Pramod
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, USA
| | - Rejwi Acharya Dahal
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, USA
| | - Michael J Tomlinson
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, USA
| | - Babita Sharma
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, USA
| | - James D Foster
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, USA
| | - Mu-Fa Zou
- Medicinal Chemistry Section, National Institute on Drug Abuse - Intramural Research Program, Baltimore, MD 21224, USA
| | - Comfort Boatang
- Medicinal Chemistry Section, National Institute on Drug Abuse - Intramural Research Program, Baltimore, MD 21224, USA
| | - Amy Hauck Newman
- Medicinal Chemistry Section, National Institute on Drug Abuse - Intramural Research Program, Baltimore, MD 21224, USA
| | - John R Lever
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, 65201, USA; Department of Radiology and Radiopharmaceutical Sciences Institute, University of Missouri, Columbia, MO 65211, USA
| | - Roxanne A Vaughan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, USA.
| | - L Keith Henry
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, USA.
| |
Collapse
|
30
|
Avelar AJ, Cao J, Newman AH, Beckstead MJ. Atypical dopamine transporter inhibitors R-modafinil and JHW 007 differentially affect D2 autoreceptor neurotransmission and the firing rate of midbrain dopamine neurons. Neuropharmacology 2017; 123:410-419. [PMID: 28625719 PMCID: PMC5546153 DOI: 10.1016/j.neuropharm.2017.06.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/17/2017] [Accepted: 06/14/2017] [Indexed: 12/12/2022]
Abstract
Abuse of psychostimulants like cocaine that inhibit dopamine (DA) reuptake through the dopamine transporter (DAT) represents a major public health issue, however FDA-approved pharmacotherapies have yet to be developed. Recently a class of ligands termed "atypical DAT inhibitors" has gained attention due to their range of effectiveness in increasing extracellular DA levels without demonstrating significant abuse liability. These compounds not only hold promise as therapeutic agents to treat stimulant use disorders but also as experimental tools to improve our understanding of DAT function. Here we used patch clamp electrophysiology in mouse brain slices to explore the effects of two atypical DAT inhibitors (R-modafinil and JHW 007) on the physiology of single DA neurons in the substantia nigra and ventral tegmental area. Despite their commonalities of being DAT inhibitors that lack cocaine-like behavioral profiles, these compounds exhibited surprisingly divergent cellular effects. Similar to cocaine, R-modafinil slowed DA neuron firing in a D2 receptor-dependent manner and rapidly enhanced the amplitude and duration of D2 receptor-mediated currents in the midbrain. In contrast, JHW 007 exhibited little effect on firing, slow DAT blockade, and an unexpected inhibition of D2 receptor-mediated currents that may be due to direct D2 receptor antagonism. Furthermore, pretreatment with JHW 007 blunted the cellular effects of cocaine, suggesting that it may be valuable to investigate similar DAT inhibitors as potential therapeutic agents. Further exploration of these and other atypical DAT inhibitors may reveal important cellular effects of compounds that will have potential as pharmacotherapies for treating cocaine use disorders.
Collapse
Affiliation(s)
- Alicia J Avelar
- Department of Cellular and Integrative Physiology, UT Health Science Center, San Antonio, TX, 78229, USA.
| | - Jianjing Cao
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA.
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA.
| | - Michael J Beckstead
- Department of Cellular and Integrative Physiology, UT Health Science Center, San Antonio, TX, 78229, USA.
| |
Collapse
|