1
|
Gu W, Gim J, Lee D, Eom H, Lee JJ, Yoon SS, Heo TY, Yun J. Artificial intelligence-based analysis of behavior and brain images in cocaine-self-administered marmosets. J Neurosci Methods 2024; 412:110294. [PMID: 39306012 DOI: 10.1016/j.jneumeth.2024.110294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 08/25/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND The sophisticated behavioral and cognitive repertoires of non-human primates (NHPs) make them suitable subjects for studies involving cocaine self-administration (SA) schedules. However, ethical considerations, adherence to the 3Rs principle (replacement, reduction and refinement), and other factors make it challenging to obtain NHPs individuals for research. Consequently, there is a need for methods that can comprehensively analyze small datasets using artificial intelligence (AI). NEW METHODS We employed AI to identify cocaine dependence patterns from collected data. First, we collected behavioral data from cocaine SA marmosets (Callithrix jacchus) to develop a dependence prediction model. SHapley Additive exPlanations (SHAP) values were used to demonstrate the importance of various variables. Additionally, we collected positron emission tomographic (PET) images showing dopamine transporter (DAT) binding potential and developed an algorithm for PET image segmentation. RESULTS The prediction model indicated that the Random Forest (RF) algorithm performed best, with an area under the curve (AUC) of 0.92. The top five variables influencing the model were identified using SHAP values. The PET image segmentation model achieved an accuracy of 0.97, a mean squared error of 0.02, an intersection over union (IoU) of 0.845, and a Dice coefficient of 0.913. COMPARISON WITH EXISTING METHODS AND CONCLUSION Utilizing data from the marmoset SA experiment, we developed an ML-based dependence prediction model and analyzed variable importance rankings using SHAP. AI-based imaging segmentation methods offer a valuable tool for evaluating DAT availability in NHPs following chronic cocaine administration.
Collapse
Affiliation(s)
- Wonmi Gu
- College of Pharmacy, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea
| | - Juhui Gim
- College of Pharmacy, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea
| | - Dohyun Lee
- Non-clinical Center, Osong Medical Innovation Foundation, 123 Osongsaengmyeong-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea
| | - Heejong Eom
- Non-clinical Center, Osong Medical Innovation Foundation, 123 Osongsaengmyeong-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea
| | - Jae Jun Lee
- Non-clinical Center, Osong Medical Innovation Foundation, 123 Osongsaengmyeong-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea
| | - Seong Shoon Yoon
- College of Korean Medicine, Daegu Haany University, 136 Sincheondong-ro, Suseong-gu, Daegu 42158, Republic of Korea
| | - Tae-Young Heo
- Department of Information & Statistics, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju-si, Chungcheongbuk-do 28644, Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea.
| |
Collapse
|
2
|
Sakayori T, Ikeda Y, Arakawa R, Nogami T, Tateno A. A randomized placebo controlled trial demonstrates the effect of dl-methylephedrine on brain functions is weaker than that of pseudoephedrine. Sci Rep 2024; 14:20793. [PMID: 39242643 PMCID: PMC11379680 DOI: 10.1038/s41598-024-71851-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024] Open
Abstract
Intellectual drug doping in athletics by using stimulants that affect central nervous system functions has been diversified. Stimulants are regulated by the World Anti-Doping Agency according to their levels of urinary concentration. Positron emission tomography could evaluate how stimulants affect central nervous system functions. We aimed to evaluate the effect of stimulants on brain function by examining the difference in brain dopamine transporter occupancy by PET after administration of dl-methylephedrine or pseudoephedrine at the clinical maximum daily dose. Four PET scans without and with drug administration (placebo, dl-methylephedrine 150 mg and pseudoephedrine 240 mg) were performed. The concentrations of dl-methylephedrine and pseudoephedrine in plasma and urine were measured. DAT occupancies in the striatum with placebo, dl-methylephedrine and pseudoephedrine were calculated by PET images. The urinary concentration of dl-methylephedrine (12.7 µg/mL) exceeded the prohibited concentration (10 µg/mL), but the DAT occupancy with dl-methylephedrine (6.1%) did not differ (p = 0.92) from that with placebo (6.2%). By contrast, although the urinary concentration of pseudoephedrine (144.8 µg/mL) was below the prohibited concentration (150 μg/mL), DAT occupancy with pseudoephedrine was 18.4%, which was higher than that with placebo (p = 0.009). At the maximum clinical dose, dl-methylephedrine was shown to have weaker effects on brain function than pseudoephedrine.
Collapse
Affiliation(s)
- Takeshi Sakayori
- Department of Neuropsychiatry, Graduate School of Medicine, Nippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo, 113-8603, Japan
| | - Yumiko Ikeda
- Department of Pharmacology, Nippon Medical School, Tokyo, Japan
| | - Ryosuke Arakawa
- Department of Pharmacology, Nippon Medical School, Tokyo, Japan
| | - Tsuyoshi Nogami
- Department of Neuropsychiatry, Graduate School of Medicine, Nippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo, 113-8603, Japan
| | - Amane Tateno
- Department of Neuropsychiatry, Graduate School of Medicine, Nippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo, 113-8603, Japan.
| |
Collapse
|
3
|
Peck EG, Holleran KM, Curry AM, Holter KM, Estave PM, Sens JP, Locke JL, Ortelli OA, George BE, Dawes MH, West AM, Alexander NJ, Kiraly DD, Farris SP, Gould RW, McCool BA, Jones SR. Synaptogyrin-3 Prevents Cocaine Addiction and Dopamine Deficits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.27.605436. [PMID: 39211138 PMCID: PMC11361146 DOI: 10.1101/2024.07.27.605436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Synaptogyrin-3, a functionally obscure synaptic vesicle protein, interacts with vesicular monoamine and dopamine transporters, bringing together dopamine release and reuptake sites. Synaptogyrin-3 was reduced by chronic cocaine exposure in both humans and rats, and synaptogyrin-3 levels inversely correlated with motivation to take cocaine in rats. Synaptogyrin-3 overexpression in dopamine neurons reduced cocaine self-administration, decreased anxiety-like behavior, and enhanced cognitive flexibility. Overexpression also enhanced nucleus accumbens dopamine signaling and prevented cocaine-induced deficits, suggesting a putative therapeutic role for synaptogyrin-3 in cocaine use disorder.
Collapse
|
4
|
Jones JD, Arout CA, Luba R, Murugesan D, Madera G, Gorsuch L, Schusterman R, Martinez S. The influence of drug class on reward in substance use disorders. Pharmacol Biochem Behav 2024; 240:173771. [PMID: 38670466 PMCID: PMC11162950 DOI: 10.1016/j.pbb.2024.173771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/26/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024]
Abstract
In the United States, the societal costs associated with drug use surpass $500 billion annually. The rewarding and reinforcing properties that drive the use of these addictive substances are typically examined concerning the neurobiological effects responsible for their abuse potential. In this review, terms such as "abuse potential," "drug," and "addictive properties" are used due to their relevance to the methodological, theoretical, and conceptual framework for understanding the phenomenon of drug-taking behavior and the associated body of preclinical and clinical literature. The use of these terms is not intended to cast aspersions on individuals with substance use disorders (SUD). Understanding what motivates substance use has been a focus of SUD research for decades. Much of this corpus of work has focused on the shared effects of each drug class to increase dopaminergic transmission within the central reward pathways of the brain, or the "reward center." However, the precise influence of each drug class on dopamine signaling, and the extent thereof, differs considerably. Furthermore, the aforementioned substances have effects on several neurobiological targets that mediate and modulate their addictive properties. The current manuscript sought to review the influence of drug class on the rewarding effects of each of the major pharmacological classes of addictive drugs (i.e., psychostimulants, opioids, nicotine, alcohol, and cannabinoids). Our review suggests that even subtle differences in drug effects can result in significant variability in the subjective experience of the drug, altering rewarding and other reinforcing effects. Additionally, this review will argue that reward (i.e., the attractive and motivational property of a stimulus) alone is not sufficient to explain the abuse liability of these substances. Instead, abuse potential is best examined as a function of both positive and negative reinforcing drug effects (i.e., stimuli that the subject will work to attain and stimuli that the subject will work to end or avoid, respectively). Though reward is central to drug use, the factors that motivate and maintain drug taking are varied and complex, with much to be elucidated.
Collapse
Affiliation(s)
- Jermaine D Jones
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Drive, New York, NY 10032, USA.
| | - Caroline A Arout
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Drive, New York, NY 10032, USA
| | - Rachel Luba
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Drive, New York, NY 10032, USA
| | - Dillon Murugesan
- CUNY School of Medicine, 160 Convent Avenue, New York, NY 10031, USA
| | - Gabriela Madera
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Drive, New York, NY 10032, USA
| | - Liam Gorsuch
- Department of Psychiatry, The University of British Columbia, 430-5950 University Blvd., Vancouver V6T 1Z3, BC, Canada
| | - Rebecca Schusterman
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Drive, New York, NY 10032, USA
| | - Suky Martinez
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Drive, New York, NY 10032, USA
| |
Collapse
|
5
|
Lee KH, Camacho-Hernandez GA, Newman AH, Shi L. The Structural Basis of the Activity Cliff in Modafinil-Based Dopamine Transporter Inhibitors. Biomolecules 2024; 14:713. [PMID: 38927116 PMCID: PMC11202288 DOI: 10.3390/biom14060713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/01/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Modafinil analogs with either a sulfoxide or sulfide moiety have improved binding affinities at the human dopamine transporter (hDAT) compared to modafinil, with lead sulfoxide-substituted analogs showing characteristics of atypical inhibition (e.g., JJC8-091). Interestingly, the only distinction between sulfoxide and sulfide substitution is the presence of one additional oxygen atom. To elucidate why such a subtle difference in ligand structure can result in different typical or atypical profiles, we investigated two pairs of analogs. Our quantum mechanical calculations revealed a more negatively charged distribution of the electrostatic potential surface of the sulfoxide substitution. Using molecular dynamics simulations, we demonstrated that sulfoxide-substituted modafinil analogs have a propensity to attract more water into the binding pocket. They also exhibited a tendency to dissociate from Asp79 and form a new interaction with Asp421, consequently promoting an inward-facing conformation of hDAT. In contrast, sulfide-substituted analogs did not display these effects. These findings elucidate the structural basis of the activity cliff observed with modafinil analogs and also enhance our understanding of the functionally relevant conformational spectrum of hDAT.
Collapse
Affiliation(s)
| | | | | | - Lei Shi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse–Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA; (K.-H.L.); (G.A.C.-H.); (A.H.N.)
| |
Collapse
|
6
|
Sung C, Oh SJ, Kim JS. Imaging Procedure and Clinical Studies of [ 18F]FP-CIT PET. Nucl Med Mol Imaging 2024; 58:185-202. [PMID: 38932763 PMCID: PMC11196481 DOI: 10.1007/s13139-024-00840-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/15/2023] [Accepted: 01/05/2024] [Indexed: 06/28/2024] Open
Abstract
N-3-[18F]fluoropropyl-2β-carbomethoxy-3β-4-iodophenyl nortropane ([18F]FP-CIT) is a radiopharmaceutical for dopamine transporter (DAT) imaging using positron emission tomography (PET) to detect dopaminergic neuronal degeneration in patients with parkinsonian syndrome. [18F]FP-CIT was granted approval by the Ministry of Food and Drug Safety in 2008 as the inaugural radiopharmaceutical for PET imaging, and it has found extensive utilization across numerous institutions in Korea. This review article presents an imaging procedure for [18F]FP-CIT PET to aid nuclear medicine physicians in clinical practice and systematically reviews the clinical studies associated with [18F]FP-CIT PET.
Collapse
Affiliation(s)
- Changhwan Sung
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505 Republic of Korea
| | - Seung Jun Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505 Republic of Korea
| | - Jae Seung Kim
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505 Republic of Korea
| |
Collapse
|
7
|
Estave PM, Albertson SE, Karkhanis AN, Jones SR. Co-targeting the kappa opioid receptor and dopamine transporter reduces motivation to self-administer cocaine and partially reverses dopamine system dysregulation. Sci Rep 2024; 14:6509. [PMID: 38499566 PMCID: PMC10948819 DOI: 10.1038/s41598-024-53463-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/31/2024] [Indexed: 03/20/2024] Open
Abstract
Cocaine disrupts dopamine (DA) and kappa opioid receptor (KOR) system activity, with long-term exposure reducing inhibiton of DA uptake by cocaine and increasing KOR system function. Single treatment therapies have not been successful for cocaine use disorder; therefore, this study focuses on a combination therapy targeting the dopamine transporter (DAT) and KOR. Sprague Dawley rats self-administered 5 days of cocaine (1.5 mg/kg/inf, max 40 inf/day, FR1), followed by 14 days on a progressive ratio (PR) schedule (0.19 mg/kg/infusion). Behavioral effects of individual and combined administration of phenmetrazine and nBNI were then examined using PR. Additionally, ex vivo fast scan cyclic voltammetry was then used to assess alterations in DA and KOR system activity in the nucleus accumbens before and after treatments. Chronic administration of phenmetrazine as well as the combination of phenmetrazine and nBNI-but not nBNI alone-significantly reduced PR breakpoints. In addition, the combination of phenmetrazine and nBNI partially reversed cocaine-induced neurodysregulations of the KOR and DA systems, indicating therapeutic benefits of targeting the DA and KOR systems in tandem. These data highlight the potential benefits of the DAT and KOR as dual-cellular targets to reduce motivation to administer cocaine and reverse cocaine-induced alterations of the DA system.
Collapse
Affiliation(s)
- Paige M Estave
- Department of Physiology and Pharmacology, Wake University Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC, 27157, USA
| | - Steven E Albertson
- Department of Physiology and Pharmacology, Wake University Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC, 27157, USA
| | - Anushree N Karkhanis
- Department of Psychology, Binghamton University - State University of New York, Binghamton, NY, 13902, USA
| | - Sara R Jones
- Department of Physiology and Pharmacology, Wake University Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC, 27157, USA.
| |
Collapse
|
8
|
Colón Ortiz R, Knerler S, Fridman LB, Mercado A, Price AS, Rosado-Franco JJ, Wilkins H, Flores BR, Orsburn BC, Williams DW. Cocaine regulates antiretroviral therapy CNS access through pregnane-x receptor-mediated drug transporter and metabolizing enzyme modulation at the blood brain barrier. Fluids Barriers CNS 2024; 21:5. [PMID: 38200564 PMCID: PMC10777548 DOI: 10.1186/s12987-023-00507-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Appropriate interactions between antiretroviral therapies (ART) and drug transporters and metabolizing enzymes at the blood brain barrier (BBB) are critical to ensure adequate dosing of the brain to achieve HIV suppression. These proteins are modulated by demographic and lifestyle factors, including substance use. While understudied, illicit substances share drug transport and metabolism pathways with ART, increasing the potential for adverse drug:drug interactions. This is particularly important when considering the brain as it is relatively undertreated compared to peripheral organs and is vulnerable to substance use-mediated damage. METHODS We used an in vitro model of the human BBB to determine the extravasation of three first-line ART drugs, emtricitabine (FTC), tenofovir (TFV), and dolutegravir (DTG), in the presence and absence of cocaine, which served as our illicit substance model. The impact of cocaine on BBB integrity and permeability, drug transporters, metabolizing enzymes, and their master transcriptional regulators were evaluated to determine the mechanisms by which substance use impacted ART central nervous system (CNS) availability. RESULTS We determined that cocaine had a selective impact on ART extravasation, where it increased FTC's ability to cross the BBB while decreasing TFV. DTG concentrations that passed the BBB were below quantifiable limits. Interestingly, the potent neuroinflammatory modulator, lipopolysaccharide, had no effect on ART transport, suggesting a specificity for cocaine. Unexpectedly, cocaine did not breach the BBB, as permeability to albumin and 4 kDa FITC-dextran, as well as tight junction proteins and adhesion molecules remained unchanged. Rather, cocaine selectively decreased the pregnane-x receptor (PXR), but not constitutive androstane receptor (CAR). Consequently, drug transporter expression and activity decreased in endothelial cells of the BBB, including p-glycoprotein (P-gp), breast cancer resistance protein (BCRP), and multidrug resistance-associated protein 4 (MRP4). Further, cytochrome P450 3A4 (CYP3A4) enzymatic activity increased following cocaine treatment that coincided with decreased expression. Finally, cocaine modulated adenylate kinases that are required to facilitate biotransformation of ART prodrugs to their phosphorylated, pharmacologically active counterparts. CONCLUSION Our findings indicate that additional considerations are needed in CNS HIV treatment strategies for people who use cocaine, as it may limit ART efficacy through regulation of drug transport and metabolizing pathways at the BBB.
Collapse
Affiliation(s)
- Rodnie Colón Ortiz
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Stephen Knerler
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Lisa B Fridman
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Alicia Mercado
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Amira-Storm Price
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Jose J Rosado-Franco
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Hannah Wilkins
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Bianca R Flores
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Benjamin C Orsburn
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Dionna W Williams
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Department of Molecular Microbiology & Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA.
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Rollins Research Center, 1510 Clifton Road NE, 30322, Atlanta, Georgia.
| |
Collapse
|
9
|
González-Saiz F, Trujols J, Vergara-Moragues E. Cocaine Effect Expectancies among Patients with Cocaine Use Disorder with and without Adult Attention Deficit Hyperactivity Disorder: Are There Any Relevant Differences? J Psychoactive Drugs 2024; 56:76-87. [PMID: 36480506 DOI: 10.1080/02791072.2022.2151951] [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] [Received: 04/25/2022] [Revised: 09/05/2022] [Accepted: 09/23/2022] [Indexed: 12/14/2022]
Abstract
Cocaine use could be modulated by drug expectancies based on previously experienced subjective effects. Some patients perceive a paradoxical calming effect after cocaine use. This study was performed to explore cocaine effect expectancies in patients diagnosed with cocaine use disorder, with and without co-occurring adult attention deficit hyperactivity disorder (ADHD). Secondly, we sought to empirically determine the presence of this paradoxical calming effect after cocaine use in patients with co-occurring adult ADHD to identify the individuals most at risk of cocaine use and relapse. Cross-sectional study using a consecutive sampling method of patients diagnosed with cocaine use disorder (n = 221) treated at public therapeutic communities in Andalusia (Spain). Participants completed a battery of instruments to assess the following variables: cocaine effect expectancies, paradoxical calming effect, adult ADHD, and other co-occurring psychiatric disorders. A multivariate binary logistic regression analysis showed that two variables, the paradoxical calming effect and antisocial personality disorder (ASPD), were independently associated with the probability of being diagnosed with adult ADHD (OR = 3.43, 95% CI = 1.88-6.26 and OR = 3.42, 95% CI = 1.30-8.95, respectively). The presence of a paradoxical calming reaction to cocaine and/or a diagnosis of ASPD in patients with cocaine use disorder increases the diagnostic suspicion of co-occurring adult ADHD.
Collapse
Affiliation(s)
- Francisco González-Saiz
- Community Mental Health Unit of Villamartin, Hospital Universitario de Jerez, Cádiz, Spain
- Department of Neuroscience, Area of Psychiatry, University of Cadiz, Cadiz, Spain
- CIBERSAM (Network of Biomedical Research Centres for Mental Health), Madrid, Spain
| | - Joan Trujols
- CIBERSAM (Network of Biomedical Research Centres for Mental Health), Madrid, Spain
- Addictive Behaviours Unit, Department of Psychiatry, Hospital de la Santa Creu I Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Esperanza Vergara-Moragues
- Department of Psychobiology and Behavioural Science. Complutense University of Madrid (UCM), Madrid, Spain
| |
Collapse
|
10
|
Estrin DJ, Kulik JM, Beacher NJ, Pawlak AP, Klein SD, West MO. Acquired Alterations in Nucleus Accumbens Responsiveness to a Cocaine-Paired Discriminative Stimulus Preceding Rats' Daily Cocaine Consumption. ADDICTION NEUROSCIENCE 2023; 8:100121. [PMID: 37664217 PMCID: PMC10470667 DOI: 10.1016/j.addicn.2023.100121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Resumption of drug taking is a primary focus for substance use disorder research and can be triggered by drug-associated environmental stimuli. The Nucleus Accumbens (NAc) is a key brain region which guides motivated behavior and is implicated in resumption. There remains a pressing need to characterize NAc neurons' responsiveness to drug associated stimuli during withdrawal and abstinence. We recorded discriminative stimulus (DS) induced NAc activity via in vivo single-unit electrophysiology in rats that self-administered cocaine. Male and female rats implanted with a jugular catheter and a microwire array in NAc Core and Shell self-administered cocaine under control of a 30s auditory DS for 6 hours per session across 14 consecutive days. Rats acquired tone discrimination within 4 sessions. To exclude pharmacological effects of circulating cocaine from all neural analyses, we studied changes in DS-induced firing only for trials preceding the first infusion of cocaine in each of the 14 sessions, which were defined as "pre-drug trials." NAc neuron responses were assessed prior to tone-evoked movement onset. Responsiveness to the DS tone was exhibited throughout all sessions by the NAc Core population, but only during Early sessions by the NAc Shell population. Both Core and Shell responded selectively to the DS, i.e., more strongly on drug taking trials, or Hits, than on Missed opportunities. These findings suggest that NAc Core and Shell play distinct roles in initiating cocaine seeking prior to daily cocaine consumption, and align with reports suggesting that as drug use becomes chronic, cue-evoked activity shifts from NAc Shell to NAc Core.
Collapse
Affiliation(s)
- David J. Estrin
- Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, 413 East 69 Street, New York, NY 10021
- Department of Psychology, Rutgers University, 152 Frelinghuysen Road, Piscataway, NJ 08854
| | - Julianna M. Kulik
- Department of Psychology, Rutgers University, 152 Frelinghuysen Road, Piscataway, NJ 08854
| | - Nicholas J. Beacher
- Department of Psychology, Rutgers University, 152 Frelinghuysen Road, Piscataway, NJ 08854
- Neural Engineering Section, National Institute on Drug Abuse, 333 Cassell Drive, Baltimore, MD 21224
| | - Anthony P. Pawlak
- Department of Psychology, Rutgers University, 152 Frelinghuysen Road, Piscataway, NJ 08854
- Center of Alcohol & Substance Use Studies, University of Minnesota-Twin Cities, 75 East River Road, Minneapolis, MN 55455
- Graduate School of Applied & Professional Psychology, University of Minnesota-Twin Cities, 75 East River Road, Minneapolis, MN 55455
| | - Samuel D. Klein
- Department of Psychology, Rutgers University, 152 Frelinghuysen Road, Piscataway, NJ 08854
- Department of Psychology, University of Minnesota-Twin Cities, 75 East River Road, Minneapolis, MN 55455
| | - Mark O. West
- Department of Psychology, Rutgers University, 152 Frelinghuysen Road, Piscataway, NJ 08854
| |
Collapse
|
11
|
Pitton Rissardo J, Caprara ALF. Neuroimaging Techniques in Differentiating Parkinson's Disease from Drug-Induced Parkinsonism: A Comprehensive Review. Clin Pract 2023; 13:1427-1448. [PMID: 37987429 PMCID: PMC10660852 DOI: 10.3390/clinpract13060128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/19/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023] Open
Abstract
Neuroimaging can provide significant benefits in evaluating patients with movement disorders associated with drugs. This literature review describes neuroimaging techniques performed to distinguish Parkinson's disease from drug-induced parkinsonism. The dopaminergic radiotracers already reported to assess patients with drug-induced parkinsonism are [123I]-FP-CIT, [123I]-β-CIT, [99mTc]-TRODAT-1, [18F]-DOPA, [18F]-AV-133, and [18F]-FP-CIT. The most studied one and the one with the highest number of publications is [123I]-FP-CIT. Fludeoxyglucose (18F) revealed a specific pattern that could predict individuals susceptible to developing drug-induced parkinsonism. Another scintigraphy method is [123I]-MIBG cardiac imaging, in which a relationship between abnormal cardiac imaging and normal dopamine transporter imaging was associated with a progression to degenerative disease in individuals with drug-induced parkinsonism. Structural brain magnetic resonance imaging can be used to assess the striatal region. A transcranial ultrasound is a non-invasive method with significant benefits regarding costs and availability. Optic coherence tomography only showed abnormalities in the late phase of Parkinson's disease, so no benefit in distinguishing early-phase Parkinson's disease and drug-induced parkinsonism was found. Most methods demonstrated a high specificity in differentiating degenerative from non-degenerative conditions, but the sensitivity widely varied in the studies. An algorithm was designed based on clinical manifestations, neuroimaging, and drug dose adjustment to assist in the management of patients with drug-induced parkinsonism.
Collapse
|
12
|
Fridman LB, Knerler S, Price AS, Ortiz RC, Mercado A, Wilkins H, Flores BR, Orsburn BC, Williams DW. Cocaine Regulates Antiretroviral Therapy CNS Access Through Pregnane-X Receptor-Mediated Drug Transporter and Metabolizing Enzyme Modulation at the Blood Brain Barrier. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.28.551042. [PMID: 37546800 PMCID: PMC10402182 DOI: 10.1101/2023.07.28.551042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Background Appropriate interactions between antiretroviral therapies (ART) and drug transporters and metabolizing enzymes at the blood brain barrier (BBB) are critical to ensure adequate dosing of the brain to achieve HIV suppression. These proteins are modulated by demographic and lifestyle factors, including substance use. While understudied, illicit substances share drug transport and metabolism pathways with ART, increasing the potential for adverse drug:drug interactions. This is particularly important when considering the brain as it is relatively undertreated compared to peripheral organs and is vulnerable to substance use-mediated damage. Methods We used an in vitro model of the human BBB to determine the extravasation of three first-line ART drugs, emtricitabine (FTC), tenofovir (TFV), and dolutegravir (DTG), in the presence and absence of cocaine, which served as our illicit substance model. The impact of cocaine on BBB integrity and permeability, drug transporters, metabolizing enzymes, and their master transcriptional regulators were evaluated to determine the mechanisms by which substance use impacted ART central nervous system (CNS) availability. Results We determined that cocaine had a selective impact on ART extravasation, where it increased FTC's ability to cross the BBB while decreasing TFV. DTG concentrations that passed the BBB were below quantifiable limits. Interestingly, the potent neuroinflammatory modulator, lipopolysaccharide, had no effect on ART transport, suggesting a specificity for cocaine. Unexpectedly, cocaine did not breach the BBB, as permeability to albumin and tight junction proteins and adhesion molecules remained unchanged. Rather, cocaine selectively decreased the pregnane-x receptor (PXR), but not constitutive androstane receptor (CAR). Consequently, drug transporter expression and activity decreased in endothelial cells of the BBB, including p-glycoprotein (P-gp), breast cancer resistance protein (BCRP), and multidrug resistance-associated protein 4 (MRP4). Further, cytochrome P450 3A4 (CYP3A4) enzymatic activity increased following cocaine treatment that coincided with decreased expression. Finally, cocaine modulated adenylate kinases are required to facilitate biotransformation of ART prodrugs to their phosphorylated, pharmacologically active counterparts. Conclusion Our findings indicate that additional considerations are needed in CNS HIV treatment strategies for people who use cocaine, as it may limit ART efficacy through regulation of drug transport and metabolizing pathways at the BBB.
Collapse
Affiliation(s)
- Lisa B. Fridman
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Stephen Knerler
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Amira-Storm Price
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Rodnie Colón Ortiz
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Alicia Mercado
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Hannah Wilkins
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Bianca R. Flores
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Benjamin C. Orsburn
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Dionna W. Williams
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
- Department of Molecular Microbiology & Immunology, Johns Hopkins School of Public Health, Baltimore, Maryland 21205
| |
Collapse
|
13
|
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 DOI: 10.1038/s41398-023-02493-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
14
|
Chahid Y, Sheikh ZH, Mitropoulos M, Booij J. A systematic review of the potential effects of medications and drugs of abuse on dopamine transporter imaging using [ 123I]I-FP-CIT SPECT in routine practice. Eur J Nucl Med Mol Imaging 2023; 50:1974-1987. [PMID: 36847827 PMCID: PMC10199883 DOI: 10.1007/s00259-023-06171-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/18/2023] [Indexed: 03/01/2023]
Abstract
PURPOSE In routine practice, dopamine transporter (DAT) imaging is frequently used as a diagnostic tool to support the diagnosis of Parkinson's disease or dementia with Lewy bodies. In 2008, we published a review on which medications and drugs of abuse may influence striatal [123I]I-FP-CIT binding and consequently may influence the visual read of an [123I]I-FP-CIT SPECT scan. We made recommendations on which drugs should be withdrawn before performing DAT imaging in routine practice. Here, we provide an update of the original work based on published research since 2008. METHODS We performed a systematic review of literature without language restriction from January 2008 until November 2022 to evaluate the possible effects of medications and drugs of abuse, including the use of tobacco and alcohol, on striatal DAT binding in humans. RESULTS The systematic literature search identified 838 unique publications, of which 44 clinical studies were selected. Using this approach, we found additional evidence to support our original recommendations as well as some new findings on potential effect of other medications on striatal DAT binding. Consequently, we updated the list of medications and drugs of abuse that may influence the visual read of [123I]I-FP-CIT SPECT scans in routine clinical practice. CONCLUSION We expect that a timely withdrawal of these medications and drugs of abuse before DAT imaging may reduce the incidence of false-positive reporting. Nevertheless, the decision to withdraw any medication must be made by the specialist in charge of the patient's care and considering the pros and cons of doing so.
Collapse
Affiliation(s)
- Youssef Chahid
- Amsterdam UMC location University of Amsterdam, Radiology and Nuclear Medicine, Meibergdreef 9, Amsterdam, The Netherlands.
- Amsterdam UMC location University of Amsterdam, Clinical Pharmacy, Meibergdreef 9, Amsterdam, The Netherlands.
| | - Zulfiqar H Sheikh
- GE Healthcare, Pharmaceutical Diagnostics, Nightingales Ln, Chalfont Saint Giles, United Kingdom
| | - Max Mitropoulos
- GE Healthcare, Pharmaceutical Diagnostics, Nightingales Ln, Chalfont Saint Giles, United Kingdom
| | - Jan Booij
- Amsterdam UMC location University of Amsterdam, Radiology and Nuclear Medicine, Meibergdreef 9, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Emerson SD, Chevée M, Mews P, Calipari ES. The transcriptional response to acute cocaine is inverted in male mice with a history of cocaine self-administration and withdrawal throughout the mesocorticolimbic system. Mol Cell Neurosci 2023; 125:103823. [PMID: 36868542 PMCID: PMC10247534 DOI: 10.1016/j.mcn.2023.103823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/30/2023] [Accepted: 02/14/2023] [Indexed: 03/05/2023] Open
Abstract
A large body of work has demonstrated that cocaine-induced changes in transcriptional regulation play a central role in the onset and maintenance of cocaine use disorder. An underappreciated aspect of this area of research, however, is that the pharmacodynamic properties of cocaine can change depending on an organism's previous drug-exposure history. In this study, we utilized RNA sequencing to characterize how the transcriptome-wide effects of acute cocaine exposure were altered by a history of cocaine self-administration and long-term withdrawal (30 days) in the ventral tegmental area (VTA), nucleus accumbens (NAc), and prefrontal cortex (PFC) in male mice. First, we found that the gene expression patterns induced by a single cocaine injection (10 mg/kg) were discordant between cocaine-naïve mice and mice in withdrawal from cocaine self-administration. Specifically, the same genes that were upregulated by acute cocaine in cocaine-naïve mice were downregulated by the same dose of cocaine in mice undergoing long-term withdrawal; the same pattern of opposite regulation was observed for the genes downregulated by initial acute cocaine exposure. When we analyzed this dataset further, we found that the gene expression patterns that were induced by long-term withdrawal from cocaine self-administration showed a high degree of overlap with the gene expression patterns of acute cocaine exposure - even though animals had not consumed cocaine in 30 days. Interestingly, cocaine re-exposure at this withdrawal time point reversed this expression pattern. Finally, we found that this pattern was similar across the VTA, PFC, NAc, and within each brain region the same genes were induced by acute cocaine, re-induced during long-term withdrawal, and reversed by cocaine re-exposure. Together, we identified a longitudinal pattern of gene regulation that is conserved across the VTA, PFC, and NAc, and characterized the genes constituting this pattern in each brain region.
Collapse
Affiliation(s)
- Soren D Emerson
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
| | - Maxime Chevée
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
| | - Philipp Mews
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Erin S Calipari
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Department of Psychiatry and Behavioral Sciences, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
16
|
Hall N, Dao N, Hewett C, Oberle S, Minagar A, Lamon K, Ford C, Blough BE, Alexander JS, Murnane KS. Methamphetamine and Designer Stimulants Modulate Tonic Human Cerebrovascular Smooth Muscle Contractility: Relevance to Drug-Induced Neurovascular Stress. PATHOPHYSIOLOGY 2023; 30:144-154. [PMID: 37092527 PMCID: PMC10123609 DOI: 10.3390/pathophysiology30020013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/28/2023] [Accepted: 04/07/2023] [Indexed: 04/25/2023] Open
Abstract
To avoid criminal prosecution, clandestine chemists produce designer stimulants that mimic the pharmacological and psychoactive effects of conventional stimulants, such as methamphetamine. Following persistent or high-dose exposure, both acute vasoconstriction and loss of vascular homeostasis are reported dangers of conventional stimulants, and designer stimulants may pose even greater dangers. To compare the effects of a conventional stimulant and two designer stimulants on vascular contraction, this study examined the direct effects of 1,3-benzodioxolylbutanamine (BDB) and N-butylpentylone in comparison to methamphetamine on the function of human brain vascular smooth muscle cells (HBVSMCs). HBVSMCs suspended in collagen gels were exposed to varying concentrations of each drug, and the degree of constriction was assessed over one week. The MTT assay was used to measure the impact of the three drugs on the cellular metabolic activity as a marker of cellular toxicity. The highest concentration tested of either methamphetamine or N-butylpentylone produced a loss of HBVSMC contractility and impaired cellular metabolism. BDB showed a similar pattern of effects, but, uniquely, it also induced vasoconstrictive effects at substantially lower concentrations. Each drug produced direct effects on HBVSMC contraction that may be a mechanism by which the cardiovascular system is damaged following high-dose or persistent exposure, and this could be exacerbated by any sympathomimetic effects of these compounds in whole organisms. BDB appears to impact HBVSMC function in ways distinct from methamphetamine and N-butylpentylone, which may present unique dangers.
Collapse
Affiliation(s)
- Nicole Hall
- Louisiana Addiction Research Center, LSU Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Nhi Dao
- Louisiana Addiction Research Center, LSU Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Caddo Parish Magnet High School, Shreveport, LA 71101, USA
| | - Cameron Hewett
- Louisiana Addiction Research Center, LSU Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Sara Oberle
- Louisiana Addiction Research Center, LSU Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Caddo Parish Magnet High School, Shreveport, LA 71101, USA
| | - Andrew Minagar
- Louisiana Addiction Research Center, LSU Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Caddo Parish Magnet High School, Shreveport, LA 71101, USA
| | - Kariann Lamon
- Louisiana Addiction Research Center, LSU Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Carey Ford
- Department of Molecular & Cellular Physiology, LSU Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Bruce E. Blough
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC 27709, USA
| | - J. Steven Alexander
- Louisiana Addiction Research Center, LSU Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Department of Molecular & Cellular Physiology, LSU Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Kevin S. Murnane
- Louisiana Addiction Research Center, LSU Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
- Department of Psychiatry & Behavioral Medicine, LSU Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| |
Collapse
|
17
|
Towers EB, Williams IL, Qillawala EI, Rissman EF, Lynch WJ. Sex/Gender Differences in the Time-Course for the Development of Substance Use Disorder: A Focus on the Telescoping Effect. Pharmacol Rev 2023; 75:217-249. [PMID: 36781217 PMCID: PMC9969523 DOI: 10.1124/pharmrev.121.000361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 10/05/2022] [Accepted: 10/25/2022] [Indexed: 12/14/2022] Open
Abstract
Sex/gender effects have been demonstrated for multiple aspects of addiction, with one of the most commonly cited examples being the "telescoping effect" where women meet criteria and/or seek treatment of substance use disorder (SUD) after fewer years of drug use as compared with men. This phenomenon has been reported for multiple drug classes including opioids, psychostimulants, alcohol, and cannabis, as well as nonpharmacological addictions, such as gambling. However, there are some inconsistent reports that show either no difference between men and women or opposite effects and a faster course to addiction in men than women. Thus, the goals of this review are to evaluate evidence for and against the telescoping effect in women and to determine the conditions/populations for which the telescoping effect is most relevant. We also discuss evidence from preclinical studies, which strongly support the validity of the telescoping effect and show that female animals develop addiction-like features (e.g., compulsive drug use, an enhanced motivation for the drug, and enhanced drug-craving/vulnerability to relapse) more readily than male animals. We also discuss biologic factors that may contribute to the telescoping effect, such as ovarian hormones, and its neurobiological basis focusing on the mesolimbic dopamine reward pathway and the corticomesolimbic glutamatergic pathway considering the critical roles these pathways play in the rewarding/reinforcing effects of addictive drugs and SUD. We conclude with future research directions, including intervention strategies to prevent the development of SUD in women. SIGNIFICANCE STATEMENT: One of the most widely cited gender/sex differences in substance use disorder (SUD) is the "telescoping effect," which reflects an accelerated course in women versus men for the development and/or seeking treatment for SUD. This review evaluates evidence for and against a telescoping effect drawing upon data from both clinical and preclinical studies. We also discuss the contribution of biological factors and underlying neurobiological mechanisms and highlight potential targets to prevent the development of SUD in women.
Collapse
Affiliation(s)
- Eleanor Blair Towers
- Psychiatry and Neurobehavioral Sciences (E.B.T., I.L.W., E.I.Q., W.J.L.) and Medical Scientist Training Program (E.B.T.), University of Virginia, Charlottesville, Virginia, and Center for Human Health and the Environment and Program in Genetics, North Carolina State University, Raleigh, North Carolina (E.F.R.)
| | - Ivy L Williams
- Psychiatry and Neurobehavioral Sciences (E.B.T., I.L.W., E.I.Q., W.J.L.) and Medical Scientist Training Program (E.B.T.), University of Virginia, Charlottesville, Virginia, and Center for Human Health and the Environment and Program in Genetics, North Carolina State University, Raleigh, North Carolina (E.F.R.)
| | - Emaan I Qillawala
- Psychiatry and Neurobehavioral Sciences (E.B.T., I.L.W., E.I.Q., W.J.L.) and Medical Scientist Training Program (E.B.T.), University of Virginia, Charlottesville, Virginia, and Center for Human Health and the Environment and Program in Genetics, North Carolina State University, Raleigh, North Carolina (E.F.R.)
| | - Emilie F Rissman
- Psychiatry and Neurobehavioral Sciences (E.B.T., I.L.W., E.I.Q., W.J.L.) and Medical Scientist Training Program (E.B.T.), University of Virginia, Charlottesville, Virginia, and Center for Human Health and the Environment and Program in Genetics, North Carolina State University, Raleigh, North Carolina (E.F.R.)
| | - Wendy J Lynch
- Psychiatry and Neurobehavioral Sciences (E.B.T., I.L.W., E.I.Q., W.J.L.) and Medical Scientist Training Program (E.B.T.), University of Virginia, Charlottesville, Virginia, and Center for Human Health and the Environment and Program in Genetics, North Carolina State University, Raleigh, North Carolina (E.F.R.)
| |
Collapse
|
18
|
Bailey LS, Bagley JR, Wherry JD, Chesler EJ, Karkhanis A, Jentsch JD, Tarantino LM. Repeated dosing with cocaine produces strain-dependent effects on responding for conditioned reinforcement in Collaborative Cross mice. Psychopharmacology (Berl) 2023; 240:561-573. [PMID: 36239767 PMCID: PMC10083021 DOI: 10.1007/s00213-022-06256-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/02/2022] [Indexed: 11/24/2022]
Abstract
RATIONALE Cocaine use disorder (CUD) is a highly heritable form of substance use disorder, with genetic variation accounting for a substantial proportion of the risk for transitioning from recreational use to a clinically impairing addiction. With repeated exposures to cocaine, psychomotor and incentive sensitization are observed in rodents. These phenomena are thought to model behavioral changes elicited by the drug that contribute to the progression into addiction, but little is known about how genetic variation may moderate these consequences. OBJECTIVES Here, we describe the use of two Collaborative Cross (CC) recombinant inbred mouse strains that either exhibit high (CC018/UncJ) or no (CC027/GeniUncJ) psychomotor sensitization in response to cocaine to measure phenotypes related to incentive sensitization after repeated cocaine exposures; given the relationship of incentive motivation to nucleus accumbens core (NAc) dopamine release and reuptake, we also assessed these neurochemical mechanisms. METHODS Adult male and female CC018/UncJ and CC027/GeniUncJ mice underwent Pavlovian conditioning to associate a visual cue with presentation of a palatable food reward, then received five, every-other-day injections of cocaine or vehicle. Following Pavlovian re-training, they underwent testing acquisition of a new operant response for the visual cue, now serving as a conditioned reinforcer. Subsequently, electrically evoked dopamine release was assessed using fast-scan cyclic voltammetry from acute brain slices containing the NAc. RESULTS While both strains acquired the Pavlovian association, only CC018/UncJ mice showed conditioned reinforcement and incentive sensitization in response to cocaine, while CC027/GeniUncJ mice did not. Voltammetry data revealed that CC018/UncJ, compared to CC027/GeniUnc, mice exhibited higher baseline dopamine release and uptake. Moreover, chronic cocaine exposure blunted tonic and phasic dopamine release in CC018/UncJ, but not CC027/GeniUncJ, mice. CONCLUSIONS Genetic background is a moderator of cocaine-induced neuroadaptations in mesolimbic dopamine signaling, which may contribute to both psychomotor and incentive sensitization and indicate a shared biological mechanism of variation.
Collapse
Affiliation(s)
- Lauren S Bailey
- Department of Psychology, State University of New York - Binghamton University, PO Box 6000, Binghamton, NY, 13902-6000, USA
| | - Jared R Bagley
- Department of Psychology, State University of New York - Binghamton University, PO Box 6000, Binghamton, NY, 13902-6000, USA
| | - James D Wherry
- Department of Psychology, State University of New York - Binghamton University, PO Box 6000, Binghamton, NY, 13902-6000, USA
| | | | - Anushree Karkhanis
- Department of Psychology, State University of New York - Binghamton University, PO Box 6000, Binghamton, NY, 13902-6000, USA
| | - James D Jentsch
- Department of Psychology, State University of New York - Binghamton University, PO Box 6000, Binghamton, NY, 13902-6000, USA.
- The Jackson Laboratory, Bar Harbor, ME, USA.
| | - Lisa M Tarantino
- The Jackson Laboratory, Bar Harbor, ME, USA
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
19
|
Fang Y, Sun Y, Liu Y, Liu T, Hao W, Liao Y. Neurobiological mechanisms and related clinical treatment of addiction: a review. PSYCHORADIOLOGY 2022; 2:180-189. [PMID: 38665277 PMCID: PMC10917179 DOI: 10.1093/psyrad/kkac021] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 04/28/2024]
Abstract
Drug addiction or substance use disorder (SUD), has been conceptualized as a three-stage (i.e. binge/intoxication, withdrawal/negative affect, and preoccupation/anticipation/craving) recurring cycle that involves complex changes in neuroplasticity, reward, motivation, desire, stress, memory, and cognitive control, and other related brain regions and brain circuits. Neuroimaging approaches, including magnetic resonance imaging, have been key to mapping neurobiological changes correlated to complex brain regions of SUD. In this review, we highlight the neurobiological mechanisms of these three stages of addiction. The abnormal activity of the ventral tegmental, nucleus accumbens, and caudate nucleus in the binge/intoxication stage involve the reward circuit of the midbrain limbic system. The changes in the orbitofrontal cortex, dorsolateral prefrontal cortex, amygdala, and hypothalamus emotional system in the withdrawal/negative affect stage involve increases in negative emotional states, dysphoric-like effects, and stress-like responses. The dysregulation of the insula and prefrontal lobes is associated with craving in the anticipation stage. Then, we review the present treatments of SUD based on these neuroimaging findings. Finally, we conclude that SUD is a chronically relapsing disorder with complex neurobiological mechanisms and multimodal stages, of which the craving stage with high relapse rate may be the key element in treatment efficacy of SUD. Precise interventions targeting different stages of SUD and characteristics of individuals might serve as a potential therapeutic strategy for SUD.
Collapse
Affiliation(s)
- Yehong Fang
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Yunkai Sun
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Yi Liu
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Tieqiao Liu
- Department of Psychiatry & Mental Health Institute of the Second Xiangya Hospital, Central South University. National Clinical Research Center on Mental Disorders & National Technology Institute on Mental Disorders. Hunan Key Laboratory of Psychiatry and Mental Health, 139 Renmin (M) Rd, Changsha, Hunan 410011, P. R. China
| | - Wei Hao
- Department of Psychiatry & Mental Health Institute of the Second Xiangya Hospital, Central South University. National Clinical Research Center on Mental Disorders & National Technology Institute on Mental Disorders. Hunan Key Laboratory of Psychiatry and Mental Health, 139 Renmin (M) Rd, Changsha, Hunan 410011, P. R. China
| | - Yanhui Liao
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| |
Collapse
|
20
|
Engeln M, Fox ME, Chandra R, Choi EY, Nam H, Qadir H, Thomas SS, Rhodes VM, Turner MD, Herman RJ, Calarco CA, Lobo MK. Transcriptome profiling of the ventral pallidum reveals a role for pallido-thalamic neurons in cocaine reward. Mol Psychiatry 2022; 27:3980-3991. [PMID: 35764708 PMCID: PMC9722585 DOI: 10.1038/s41380-022-01668-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/28/2022] [Accepted: 06/08/2022] [Indexed: 02/06/2023]
Abstract
Psychostimulant exposure alters the activity of ventral pallidum (VP) projection neurons. However, the molecular underpinnings of these circuit dysfunctions are unclear. We used RNA-sequencing to reveal alterations in the transcriptional landscape of the VP that are induced by cocaine self-administration in mice. We then probed gene expression in select VP neuronal subpopulations to isolate a circuit associated with cocaine intake. Finally, we used both overexpression and CRISPR-mediated knockdown to test the role of a gene target on cocaine-mediated behaviors as well as dendritic spine density. Our results showed that a large proportion (55%) of genes associated with structural plasticity were changed 24 h following cocaine intake. Among them, the transcription factor Nr4a1 (Nuclear receptor subfamily 4, group A, member 1, or Nur77) showed high expression levels. We found that the VP to mediodorsal thalamus (VP → MDT) projection neurons specifically were recapitulating this increase in Nr4a1 expression. Overexpressing Nr4a1 in VP → MDT neurons enhanced drug-seeking and drug-induced reinstatement, while Nr4a1 knockdown prevented self-administration acquisition and subsequent cocaine-mediated behaviors. Moreover, we showed that Nr4a1 negatively regulated spine dynamics in this specific cell subpopulation. Together, our study identifies for the first time the transcriptional mechanisms occurring in VP in drug exposure. Our study provides further understanding on the role of Nr4a1 in cocaine-related behaviors and identifies the crucial role of the VP → MDT circuit in drug intake and relapse-like behaviors.
Collapse
Affiliation(s)
- Michel Engeln
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA.
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, F-33000, Bordeaux, France.
| | - Megan E Fox
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Anesthesiology & Perioperative Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Ramesh Chandra
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Eric Y Choi
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hyungwoo Nam
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Houman Qadir
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Shavin S Thomas
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Victoria M Rhodes
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Makeda D Turner
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rae J Herman
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Cali A Calarco
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
21
|
The impact of caloric availability on eating behavior and ultra-processed food reward. Appetite 2022; 178:106274. [PMID: 35963586 DOI: 10.1016/j.appet.2022.106274] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 07/07/2022] [Accepted: 08/07/2022] [Indexed: 12/19/2022]
Abstract
The food environment has changed rapidly and dramatically in the last 50 years. While industrial food processing has increased the safety and stability of the food supply, a rapid expansion in the scope and scale of food processing in the 1980's has resulted in a market dominated by ultra-processed foods. Here, we use the NOVA definition of category 4 ultra-processed foods (UPFs) as they make up around 58% of total calories consumed in the US and 66% of calories in US children. UPFs are formulated from ingredients with no or infrequent culinary use, contain additives, and have a long shelf-life, spending long periods in contact with packaging materials, allowing for the absorption of compounds from those materials. The full implications of this dietary shift to UPFs on human health and disease outcomes are difficult, if not impossible, to quantify. However, UPF consumption is linked with various forms of cancer, increased cardiovascular disease, and increased all-cause mortality. Understanding food choice is, therefore, a critical problem in health research. Although many factors influence food choice, here we focus on the properties of the foods themselves. UPFs are generally treated as food, not as the highly refined, industrialized substances that they are, whose properties and components must be studied. Here, we examine one property of UPFs, that they deliver useable calories rapidly as a potential factor driving UPF overconsumption. First, we explore evidence that UPFs deliver calories more rapidly. Next, we examine the role of the gut-brain axis and its interplay with canonical reward systems, and last, we describe how speed affects both basic learning processes and drugs of abuse.
Collapse
|
22
|
Müller CP. Serotonin and Consciousness-A Reappraisal. Behav Brain Res 2022; 432:113970. [PMID: 35716774 DOI: 10.1016/j.bbr.2022.113970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 11/02/2022]
Abstract
The serotonergic system of the brain is a major modulator of behaviour. Here we describe a re-appraisal of its function for consciousness based on anatomical, functional and pharmacological data. For a better understanding, the current model of consciousness is expanded. Two parallel streams of conscious flow are distinguished. A flow of conscious content and an affective consciousness flow. While conscious content flow has its functional equivalent in the activity of higher cortico-cortical and cortico-thalamic networks, affective conscious flow originates in segregated deeper brain structures for single emotions. It is hypothesized that single emotional networks converge on serotonergic and other modulatory transmitter neurons in the brainstem where a bound percept of an affective conscious flow is formed. This is then dispersed to cortical and thalamic networks, where it is time locked with conscious content flow at the level of these networks. Serotonin acts in concert with other modulatory systems of the brain stem with some possible specialization on single emotions. Together, these systems signal a bound percept of affective conscious flow. Dysfunctions in the serotonergic system may not only give rise to behavioural and somatic symptoms, but also essentially affect the coupling of conscious affective flow with conscious content flow, leading to the affect-stained subjective side of mental disorders like anxiety, depression, or schizophrenia. The present model is an attempt to integrate the growing insights into serotonergic system function. However, it is acknowledged, that several key claims are still at a heuristic level that need further empirical support.
Collapse
Affiliation(s)
- Christian P Müller
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany; Centre for Drug Research, Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia.
| |
Collapse
|
23
|
Dominic P, Ahmad J, Awwab H, Bhuiyan MS, Kevil CG, Goeders NE, Murnane KS, Patterson JC, Sandau KE, Gopinathannair R, Olshansky B. Stimulant Drugs of Abuse and Cardiac Arrhythmias. Circ Arrhythm Electrophysiol 2022; 15:e010273. [PMID: 34961335 PMCID: PMC8766923 DOI: 10.1161/circep.121.010273] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Nonmedical use of prescription and nonprescription drugs is a worldwide epidemic, rapidly growing in magnitude with deaths because of overdose and chronic use. A vast majority of these drugs are stimulants that have various effects on the cardiovascular system including the cardiac rhythm. Drugs, like cocaine and methamphetamine, have measured effects on the conduction system and through several direct and indirect pathways, utilizing multiple second messenger systems, change the structural and electrical substrate of the heart, thereby promoting cardiac dysrhythmias. Substituted amphetamines and cocaine affect the expression and activation kinetics of multiple ion channels and calcium signaling proteins resulting in EKG changes, and atrial and ventricular brady and tachyarrhythmias. Preexisting conditions cause substrate changes in the heart, which decrease the threshold for such drug-induced cardiac arrhythmias. The treatment of cardiac arrhythmias in patients who take drugs of abuse may be specialized and will require an understanding of the unique underlying mechanisms and necessitates a multidisciplinary approach. The use of primary or secondary prevention defibrillators in drug abusers with chronic systolic heart failure is both sensitive and controversial. This review provides a broad overview of cardiac arrhythmias associated with stimulant substance abuse and their management.
Collapse
Affiliation(s)
- Paari Dominic
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center-Shreveport, LA, Department of Medicine, Louisiana State University Health Sciences Center-Shreveport, LA
| | - Javaria Ahmad
- Department of Medicine, Louisiana State University Health Sciences Center-Shreveport, LA
| | - Hajra Awwab
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center-Shreveport, LA, Department of Medicine, Louisiana State University Health Sciences Center-Shreveport, LA
| | - Md. Shenuarin Bhuiyan
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center-Shreveport, LA, Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, Department of Molecular and Cellular Physiology Louisiana State University Health Sciences Center, Shreveport, LA
| | - Christopher G. Kevil
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center-Shreveport, LA, Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, Department of Molecular and Cellular Physiology Louisiana State University Health Sciences Center, Shreveport, LA, Department of Cellular Biology and Anatomy Louisiana State University Health Sciences Center, Shreveport, LA
| | - Nicholas E. Goeders
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, LA
| | - Kevin S. Murnane
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, LA, Department of Psychiatry, Louisiana State University Health Sciences Center, Shreveport, LA
| | - James C. Patterson
- Department of Psychiatry, Louisiana State University Health Sciences Center, Shreveport, LA
| | | | - Rakesh Gopinathannair
- The Kansas City Heart Rhythm Institute (KCHRI) & Research Foundation, Overland Park Regional Medical Center, Overland Park, KS
| | - Brian Olshansky
- University of Iowa Carver College of Medicine, Iowa City, IA
| |
Collapse
|
24
|
Cabé J, Brousse G, Pereira B, Cabé N, Karsinti E, Zerdazi EH, Icick R, Llorca PM, Bloch V, Vorspan F, De Chazeron I. Influence of Clinical Markers of Dopaminergic Behaviors on Depressive Symptoms During Withdrawal in Cocaine Users. Front Psychiatry 2021; 12:775670. [PMID: 34880796 PMCID: PMC8645893 DOI: 10.3389/fpsyt.2021.775670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/29/2021] [Indexed: 12/04/2022] Open
Abstract
Background: During cocaine withdrawal, transient depressive symptoms that do not meet the criteria for depression, but promote relapse, are frequently observed. Their temporality could evoke a role of dopamine, especially since the underlying mechanism of these depressive symptoms is not well understood. We hypothesized that variation in the dopaminergic activity profile, modeled from clinical markers, could be implicated in the development of depressive symptoms during cocaine withdrawal. Methods: We compared patients reporting depressive symptoms (RDS+) or not (RDS-) during cocaine withdrawal. We evaluated dopaminergic activity through indirect clinical markers based on the known dopaminergic behaviors. A combined criterion was constructed for hyper and hypo dopaminergic models according to the O'Brien method and illustrated by the Hedges' effect-size and forest-plot graph. A multidimensional factorial analysis was carried out to determine which parameters discriminate RDS+/RDS- patients. Results: 313 patients were included, and 77% reported depressive symptoms during cocaine withdrawal. Hyperdopaminergic variables used to discriminate the two groups had a large overall effect size (-0.669) and included psychotic symptoms (-0.524), hallucinations (-0.548), and delusions (-0.528). The overall effect of the hypodopaminergic component was considerable (-0.604) with a large effect size for the severity of dependence (-0.616), withdrawal symptoms (-0.578), and anhedonia (-0.528). The combined model including hyperdopaminergic and hypodopaminergic components had the largest effect size (-0.785). Conclusion: The dopaminergic activities profile, assessed by indirect clinical markers, seems to characterize patients with depressive symptoms very well during cocaine withdrawal. RDS+ patients reported moreover higher levels of psychotic symptoms and more severe cocaine use disorder than RDS-.
Collapse
Affiliation(s)
- Julien Cabé
- Service d'addictologie et pathologies duelles, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
- Université Clermont Auvergne, CHU, CNRS, Clermont Auvergne INP, Institut Pascal, Clermont-Ferrand, France
- Faculté de Médecine, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Georges Brousse
- Service d'addictologie et pathologies duelles, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
- Université Clermont Auvergne, CHU, CNRS, Clermont Auvergne INP, Institut Pascal, Clermont-Ferrand, France
- Faculté de Médecine, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Bruno Pereira
- Direction de la Recherche Clinique et des Innovations, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Nicolas Cabé
- Normandie University, UNICAEN, PSL Université de Paris, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de la Mémoire Humaine, Caen, France
- Service d'Addictologie, Centre Hospitalier Universitaire de Caen, Caen, France
| | - Emily Karsinti
- INSERM UMR-S 1144, Université de Paris, Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France
- Département de Psychiatrie et de Médecine Addictologique, Hôpital Fernand Widal, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - El-Hadi Zerdazi
- INSERM UMR-S 1144, Université de Paris, Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France
- Département de Psychiatrie et de Médecine Addictologique, Hôpital Fernand Widal, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Romain Icick
- INSERM UMR-S 1144, Université de Paris, Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France
- Département de Psychiatrie et de Médecine Addictologique, Hôpital Fernand Widal, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Pierre M. Llorca
- Université Clermont Auvergne, CHU, CNRS, Clermont Auvergne INP, Institut Pascal, Clermont-Ferrand, France
- Faculté de Médecine, Université Clermont Auvergne, Clermont-Ferrand, France
- Service de Psychiatrie B, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Vanessa Bloch
- INSERM UMR-S 1144, Université de Paris, Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France
- Département de Psychiatrie et de Médecine Addictologique, Hôpital Fernand Widal, Assistance Publique–Hôpitaux de Paris, Paris, France
- Faculté de Médecine, Université de Paris, Paris, France
| | - Florence Vorspan
- INSERM UMR-S 1144, Université de Paris, Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France
- Département de Psychiatrie et de Médecine Addictologique, Hôpital Fernand Widal, Assistance Publique–Hôpitaux de Paris, Paris, France
- Faculté de Médecine, Université de Paris, Paris, France
| | - Ingrid De Chazeron
- Service d'addictologie et pathologies duelles, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
- Université Clermont Auvergne, CHU, CNRS, Clermont Auvergne INP, Institut Pascal, Clermont-Ferrand, France
- Faculté de Médecine, Université Clermont Auvergne, Clermont-Ferrand, France
| |
Collapse
|
25
|
Synaptic Zn 2+ potentiates the effects of cocaine on striatal dopamine neurotransmission and behavior. Transl Psychiatry 2021; 11:570. [PMID: 34750356 PMCID: PMC8575899 DOI: 10.1038/s41398-021-01693-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/15/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
Abstract
Cocaine binds to the dopamine (DA) transporter (DAT) to regulate cocaine reward and seeking behavior. Zinc (Zn2+) also binds to the DAT, but the in vivo relevance of this interaction is unknown. We found that Zn2+ concentrations in postmortem brain (caudate) tissue from humans who died of cocaine overdose were significantly lower than in control subjects. Moreover, the level of striatal Zn2+ content in these subjects negatively correlated with plasma levels of benzoylecgonine, a cocaine metabolite indicative of recent use. In mice, repeated cocaine exposure increased synaptic Zn2+ concentrations in the caudate putamen (CPu) and nucleus accumbens (NAc). Cocaine-induced increases in Zn2+ were dependent on the Zn2+ transporter 3 (ZnT3), a neuronal Zn2+ transporter localized to synaptic vesicle membranes, as ZnT3 knockout (KO) mice were insensitive to cocaine-induced increases in striatal Zn2+. ZnT3 KO mice showed significantly lower electrically evoked DA release and greater DA clearance when exposed to cocaine compared to controls. ZnT3 KO mice also displayed significant reductions in cocaine locomotor sensitization, conditioned place preference (CPP), self-administration, and reinstatement compared to control mice and were insensitive to cocaine-induced increases in striatal DAT binding. Finally, dietary Zn2+ deficiency in mice resulted in decreased striatal Zn2+ content, cocaine locomotor sensitization, CPP, and striatal DAT binding. These results indicate that cocaine increases synaptic Zn2+ release and turnover/metabolism in the striatum, and that synaptically released Zn2+ potentiates the effects of cocaine on striatal DA neurotransmission and behavior and is required for cocaine-primed reinstatement. In sum, these findings reveal new insights into cocaine's pharmacological mechanism of action and suggest that Zn2+ may serve as an environmentally derived regulator of DA neurotransmission, cocaine pharmacodynamics, and vulnerability to cocaine use disorders.
Collapse
|
26
|
Oliva F, Mangiapane C, Nibbio G, Berchialla P, Colombi N, Vigna-Taglianti FD. Prevalence of cocaine use and cocaine use disorder among adult patients with attention-deficit/hyperactivity disorder: A systematic review and meta-analysis. J Psychiatr Res 2021; 143:587-598. [PMID: 33199055 DOI: 10.1016/j.jpsychires.2020.11.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/15/2022]
Abstract
We conducted this systematic review and meta-analysis (registered with PROSPERO CRD42020142039) of the literature to estimate the lifetime prevalence of cocaine use and cocaine use disorder among adult patients with attention-deficit/hyperactivity disorder (ADHD). The literature search was performed on the electronic databases PubMed and PsychINFO without date or language restrictions. Additional studies were identified by hand searching of citations. Inclusion criteria were: studies involving adult patients with ADHD and reporting cocaine use and/or cocaine use disorders. Data were pooled in the meta-analyses using a generalized linear mixed model with random effects. Statistical heterogeneity was assessed using the Cochran Q test. Sensitivity analyses were conducted. Twelve studies were included in the review: six in the meta-analysis of cocaine use and nine in the meta-analysis of cocaine use disorder. The estimated prevalence of cocaine use was 26.0% (95% CI 0.18-0.35) and the estimated prevalence of cocaine use disorder was 10.0% (95% CI 0.08-0.13). Heterogeneity in both meta-analyses was high but decreased to non-significance in the meta-analysis on cocaine use disorder after excluding the outlier study. In conclusion, one out of four adult patients with ADHD use cocaine and one out of ten develop a lifetime cocaine use disorder. Since cocaine use can lead to more severe and complex disorders of impaired systemic functioning, adult patients with ADHD should be assessed for cocaine use disorder and promptly referred for treatment.
Collapse
Affiliation(s)
- F Oliva
- Department of Clinical and Biological Sciences, University of Torino, Italy
| | - C Mangiapane
- Department of Neurosciences "Rita Levi Montalcini", University of Torino, Italy
| | - G Nibbio
- Department of Psychiatry, Brescia University School of Medicine, Brescia, Italy
| | - P Berchialla
- Department of Clinical and Biological Sciences, University of Torino, Italy
| | - N Colombi
- Federated Library of Medicine "F. Rossi", University of Torino, Italy
| | - F D Vigna-Taglianti
- Department of Clinical and Biological Sciences, University of Torino, Italy; Piedmont Centre for Drug Addiction Epidemiology, ASL TO3, Grugliasco, Torino, Italy.
| |
Collapse
|
27
|
Kappa Opioid Receptor Mediated Differential Regulation of Serotonin and Dopamine Transporters in Mood and Substance Use Disorder. Handb Exp Pharmacol 2021; 271:97-112. [PMID: 34136961 DOI: 10.1007/164_2021_499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Dynorphin (DYN) is an endogenous neurosecretory peptide which exerts its activity by binding to the family of G protein-coupled receptors, namely the kappa opioid receptor (KOR). Opioids are associated with pain, analgesia, and drug abuse, which play a central role in mood disorders with monoamine neurotransmitter interactions. Growing evidence demonstrates the cellular signaling cascades linked to KOR-mediated monoamine transporters regulation in cell models and native brain tissues. This chapter will review DYN/KOR role in mood and addiction in relevance to dopaminergic and serotonergic neurotransmissions. Also, we discuss the recent findings on KOR-mediated differential regulation of serotonin and dopamine transporters (SERT and DAT). These findings led to a better understanding of the role of DYN/KOR system in aminergic neurotransmission via its modulatory effect on both amine release and clearance. Detailed knowledge of these processes at the molecular level enables designing novel pharmacological reagents to target transporter motifs to treat mood and addiction and reduce unwanted side effects such as aversion, dysphoria, sedation, and psychomimesis.
Collapse
|
28
|
Ruchala I, Battisti UM, Nguyen VT, Chen RYT, Glennon RA, Eltit JM. Functional characterization of N-octyl 4-methylamphetamine variants and related bivalent compounds at the dopamine and serotonin transporters using Ca 2+ channels as sensors. Toxicol Appl Pharmacol 2021; 419:115513. [PMID: 33785354 PMCID: PMC8148225 DOI: 10.1016/j.taap.2021.115513] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/21/2021] [Accepted: 03/25/2021] [Indexed: 10/21/2022]
Abstract
The early characterization of ligands at the dopamine and serotonin transporters, DAT and SERT, respectively, is important for drug discovery, forensic sciences, and drug abuse research. 4-Methyl amphetamine (4-MA) is a good example of an abused drug whose overdose can be fatal. It is a potent substrate at DAT and SERT where its simplest secondary amine (N-methyl 4-MA) retains substrate activity at them. In contrast, N-n-butyl 4-MA is very weak, therefore it was categorized as inactive at these transporters. Here, N-octyl 4-MA and other related compounds were synthesized, and their activities were evaluated at DAT and SERT. To expedite this endeavor, cells expressing DAT or SERT were co-transfected with a voltage-gated Ca2+ channel and, the genetically-encoded Ca2+ sensor, GCaMP6s. Control compounds and the newly synthesized molecules were tested on these cells using an automated multi-well fluorescence plate reader; substrates and inhibitors were identified successfully at DAT and SERT. N-Octyl 4-MA and three bivalent compounds were inhibitors at these transporters. These findings were validated by measuring Ca2+-mobilization using quantitative fluorescence microscopy. The bivalent molecules were the most potent of the series and were further characterized in an uptake-inhibition assay. Compared to cocaine, they showed comparable potency inhibiting uptake at DAT and higher potency at SERT. These observations support a previous hypothesis that amphetamine-related (and, here, N-extended alkyl and) bivalent arylalkylamine molecules are active at monoamine transporters, showing potent activity as reuptake inhibitors, and implicate the involvement of a distant auxiliary binding feature to account for their actions at DAT and SERT.
Collapse
Affiliation(s)
- Iwona Ruchala
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, United States of America
| | - Umberto M Battisti
- Deparment of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, United States of America
| | - Vy T Nguyen
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, United States of America
| | - Rita Yu-Tzu Chen
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, United States of America
| | - Richard A Glennon
- Deparment of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, United States of America
| | - Jose M Eltit
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, United States of America.
| |
Collapse
|
29
|
Supramammillary neurons projecting to the septum regulate dopamine and motivation for environmental interaction in mice. Nat Commun 2021; 12:2811. [PMID: 33990558 PMCID: PMC8121914 DOI: 10.1038/s41467-021-23040-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 04/06/2021] [Indexed: 12/29/2022] Open
Abstract
The supramammillary region (SuM) is a posterior hypothalamic structure, known to regulate hippocampal theta oscillations and arousal. However, recent studies reported that the stimulation of SuM neurons with neuroactive chemicals, including substances of abuse, is reinforcing. We conducted experiments to elucidate how SuM neurons mediate such effects. Using optogenetics, we found that the excitation of SuM glutamatergic (GLU) neurons was reinforcing in mice; this effect was relayed by their projections to septal GLU neurons. SuM neurons were active during exploration and approach behavior and diminished activity during sucrose consumption. Consistently, inhibition of SuM neurons disrupted approach responses, but not sucrose consumption. Such functions are similar to those of mesolimbic dopamine neurons. Indeed, the stimulation of SuM-to-septum GLU neurons and septum-to-ventral tegmental area (VTA) GLU neurons activated mesolimbic dopamine neurons. We propose that the supramammillo-septo-VTA pathway regulates arousal that reinforces and energizes behavioral interaction with the environment.
Collapse
|
30
|
Ragu Varman D, Subler MA, Windle JJ, Jayanthi LD, Ramamoorthy S. Novelty-induced hyperactivity and suppressed cocaine induced locomotor activation in mice lacking threonine 53 phosphorylation of dopamine transporter. Behav Brain Res 2021; 408:113267. [PMID: 33794225 DOI: 10.1016/j.bbr.2021.113267] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/03/2021] [Accepted: 03/24/2021] [Indexed: 12/26/2022]
Abstract
Dopamine (DA) transporter (DAT) is dynamically regulated by several protein kinases and the Thr53 phosphorylation of DAT (pT53-DAT) is documented in heterologous cell models and in rat brain. However, the role of endogenous pT53-DAT in living animals has never been addressed. Here we generated and studied the pT53-lacking DAT mouse model (DAT-Ala53) by CRISPR/Cas9 technology. DAT-Ala53 mice showed normal growth, body weight, body temperature, grip strength, and sucrose preference while pT53-DAT was completely absent. However, DAT-Ala53 mice showed hyperlocomotion, pronounced vertical exploratory behavior, and stereotypy in a novel environment compared to wild-type littermates (WT). DAT-Ala53 mice displayed unaltered levels of monoamines, glutamate, and GABA in the striatum compared to WT. There were also no significant differences between DAT-Ala53 mice and WT in tyrosine hydroxylase (TH) and phospho-TH levels, or in total and surface DAT levels, or in DA-transport kinetic parameters Vmax and Km. Immunohistochemical and colocalization analyses of TH and DAT in caudate-putamen and nucleus accumbens revealed no significant differences between DAT-Ala53 and WT mice. Interestingly, cocaine's potency to inhibit striatal DA transport and cocaine-induced locomotor activation were significantly reduced in the DAT-Ala53 mice. Also, ERK1/2 inhibitors completely failed to inhibit striatal DA uptake in DAT-Ala53 mice. Collectively, our findings reveal that the mice lacking pT53-DAT display novelty-induced hyperactive phenotype despite having normal transporter protein expression, DA-transport kinetics and DA-linked markers. The results also reveal that the lack of endogenous pT53-DAT renders DAT resistant to ERK1/2 inhibition and also less susceptible to cocaine inhibition and cocaine-evoked locomotor stimulation.
Collapse
Affiliation(s)
- Durairaj Ragu Varman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Mark A Subler
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Jolene J Windle
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Lankupalle D Jayanthi
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Sammanda Ramamoorthy
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA.
| |
Collapse
|
31
|
Coley AA, Padilla-Coreano N, Patel R, Tye KM. Valence processing in the PFC: Reconciling circuit-level and systems-level views. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 158:171-212. [PMID: 33785145 DOI: 10.1016/bs.irn.2020.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
An essential component in animal behavior is the ability to process emotion and dissociate among positive and negative valence in response to a rewarding or aversive stimulus. The medial prefrontal cortex (mPFC)-responsible for higher order executive functions that include cognition, learning, and working memory; and is also involved in sociability-plays a major role in emotional processing and control. Although the amygdala is widely regarded as the "emotional hub," the mPFC encodes for context-specific salience and elicits top-down control over limbic circuitry. The mPFC can then conduct behavioral responses, via cortico-striatal and cortico-brainstem pathways, that correspond to emotional stimuli. Evidence shows that abnormalities within the mPFC lead to sociability deficits, working memory impairments, and drug-seeking behavior that include addiction and compulsive disorders; as well as conditions such as anhedonia. Recent studies investigate the effects of aberrant salience processing on cortical circuitry and neuronal populations associated with these behaviors. In this chapter, we discuss mPFC valence processing, neuroanatomical connections, and physiological substrates involved in mPFC-associated behavior. We review neurocomputational and theoretical models such as "mixed selectivity," that describe cognitive control, attentiveness, and motivational drives. Using this knowledge, we describe the effects of valence imbalances and its influence on mPFC neural pathways that contribute to deficits in social cognition, while understanding the effects in addiction/compulsive behaviors and anhedonia.
Collapse
Affiliation(s)
- Austin A Coley
- Salk Institute for Biological Studies, La Jolla, CA, United States
| | | | - Reesha Patel
- Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Kay M Tye
- Salk Institute for Biological Studies, La Jolla, CA, United States.
| |
Collapse
|
32
|
Nicolucci C, Pais ML, Santos AC, Ribeiro FM, Encarnação PMCC, Silva ALM, Castro IF, Correia PMM, Veloso JFCA, Reis J, Lopes MZ, Botelho MF, Pereira FC, Priolli DG. Single Low Dose of Cocaine-Structural Brain Injury Without Metabolic and Behavioral Changes. Front Neurosci 2021; 14:589897. [PMID: 33584173 PMCID: PMC7874143 DOI: 10.3389/fnins.2020.589897] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/15/2020] [Indexed: 11/13/2022] Open
Abstract
Chronic cocaine use has been shown to lead to neurotoxicity in rodents and humans, being associated with high morbidity and mortality rates. However, recreational use, which may lead to addictive behavior, is often neglected. This occurs, in part, due to the belief that exposure to low doses of cocaine comes with no brain damage risk. Cocaine addicts have shown glucose metabolism changes related to dopamine brain activity and reduced volume of striatal gray matter. This work aims to evaluate the morphological brain changes underlying metabolic and locomotor behavioral outcome, in response to a single low dose of cocaine in a pre-clinical study. In this context, a Balb-c mouse model has been chosen, and animals were injected with a single dose of cocaine (0.5 mg/kg). Control animals were injected with saline. A behavioral test, positron emission tomography (PET) imaging, and anatomopathological studies were conducted with this low dose of cocaine, to study functional, metabolic, and morphological brain changes, respectively. Animals exposed to this cocaine dose showed similar open field activity and brain metabolic activity as compared with controls. However, histological analysis showed alterations in the prefrontal cortex and hippocampus of mice exposed to cocaine. For the first time, it has been demonstrated that a single low dose of cocaine, which can cause no locomotor behavioral and brain metabolic changes, can induce structural damage. These brain changes must always be considered regardless of the dosage used. It is essential to alert the population even against the consumption of low doses of cocaine.
Collapse
Affiliation(s)
- Camilla Nicolucci
- Multidisciplinary Research Laboratory, São Francisco University Post-graduation Stricto Sensu Programme, Bragança Paulista, Brazil
| | - Mariana Lapo Pais
- Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, Institute of Biophysics, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research, University of Coimbra, Coimbra, Portugal
| | - A C Santos
- Faculty of Medicine, Institute of Biophysics, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, Coimbra, Portugal
| | - Fabiana M Ribeiro
- Department of Physics, Institute for Nanostructures, Nanomodelling and Nanofabrication (i3N), University of Aveiro, Aveiro, Portugal
| | - Pedro M C C Encarnação
- Department of Physics, Institute for Nanostructures, Nanomodelling and Nanofabrication (i3N), University of Aveiro, Aveiro, Portugal
| | - Ana L M Silva
- Department of Physics, Institute for Nanostructures, Nanomodelling and Nanofabrication (i3N), University of Aveiro, Aveiro, Portugal.,Radiation Imaging Technologies Lda, Ílhavo, Portugal
| | - I F Castro
- Radiation Imaging Technologies Lda, Ílhavo, Portugal
| | - Pedro M M Correia
- Department of Physics, Institute for Nanostructures, Nanomodelling and Nanofabrication (i3N), University of Aveiro, Aveiro, Portugal.,Radiation Imaging Technologies Lda, Ílhavo, Portugal
| | - João F C A Veloso
- Department of Physics, Institute for Nanostructures, Nanomodelling and Nanofabrication (i3N), University of Aveiro, Aveiro, Portugal
| | - Julie Reis
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research, University of Coimbra, Coimbra, Portugal
| | - Marina Z Lopes
- Multidisciplinary Research Laboratory, São Francisco University Scientific Initiation Programme, Bragança Paulista, Brazil
| | - Maria F Botelho
- Faculty of Medicine, Institute of Biophysics, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research, University of Coimbra, Coimbra, Portugal
| | - Frederico C Pereira
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.,Faculty of Medicine, Institute of Pharmacology and Experimental Therapeutics, University of Coimbra, Coimbra, Portugal
| | - Denise G Priolli
- Multidisciplinary Research Laboratory, São Francisco University Post-graduation Stricto Sensu Programme, Bragança Paulista, Brazil
| |
Collapse
|
33
|
Havlicek DF, Rosenberg JB, De BP, Hicks MJ, Sondhi D, Kaminsky SM, Crystal RG. Cocaine vaccine dAd5GNE protects against moderate daily and high-dose "binge" cocaine use. PLoS One 2020; 15:e0239780. [PMID: 33253224 PMCID: PMC7703925 DOI: 10.1371/journal.pone.0239780] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 09/14/2020] [Indexed: 01/23/2023] Open
Abstract
The cocaine vaccine dAd5GNE is comprised of a disrupted serotype 5 adenovirus gene therapy vector covalently conjugated to the cocaine analog GNE. The vaccine evokes a high titer of circulating anti-cocaine antibodies that prevent cocaine from reaching its cognate receptors in the central nervous system. Prior studies have demonstrated the efficacy of dAd5GNE in models of occasional, moderate cocaine use. However, previous studies have not sufficiently evaluated the efficacy of dAd5GNE in models of the repetitive and high-dose "binge" use patterns common in human addicts. In the present study, we evaluated the capacity of dAd5GNE vaccination to protect against "binge" cocaine use and circumstances where vaccinated addicts attempt to override the vaccine. We modeled repetitive daily cocaine use in vaccinated Balb/c mice and African green monkeys, and evaluated high-dose "binge" scenarios in Balb/c mice. In each model of daily use the dAd5GNE vaccine prevented cocaine from reaching the central nervous system. In the high-dose "binge" model, vaccination decreased cocaine-induced hyperactivity and reduced the number of cocaine-induced seizures. Based on this data and our prior data in rodents and nonhuman primates, we have initiated a clinical trial evaluating the dAd5GNE anti-cocaine vaccine as a potential therapy for cocaine addicts who wish to stop cocaine use. If dAd5GNE vaccination is safe and produces high anti-cocaine antibody titers in the clinic, we hypothesize that the vaccine will restrict the access of cocaine to the central nervous system and inhibit cocaine-induced "highs" even in the context of moderate daily and high-dose "binge" use that might otherwise cause a drug-induced overdose.
Collapse
Affiliation(s)
- David F. Havlicek
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Jonathan B. Rosenberg
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Bishnu P. De
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Martin J. Hicks
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Dolan Sondhi
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Stephen M. Kaminsky
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Ronald G. Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| |
Collapse
|
34
|
Zakiniaeiz Y, Cosgrove KP. Designing Neuroimaging Studies to Help Inform the Clinical Treatment of Addiction. Biol Psychiatry 2020; 88:741-743. [PMID: 33092691 PMCID: PMC11265605 DOI: 10.1016/j.biopsych.2020.08.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 08/14/2020] [Accepted: 08/15/2020] [Indexed: 02/04/2023]
Affiliation(s)
- Yasmin Zakiniaeiz
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut; Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut
| | - Kelly P Cosgrove
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut; Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut; Department of Psychiatry, Yale University, New Haven, Connecticut.
| |
Collapse
|
35
|
Müller CP. Drug instrumentalization. Behav Brain Res 2020; 390:112672. [PMID: 32442549 DOI: 10.1016/j.bbr.2020.112672] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/19/2022]
Abstract
Psychoactive drugs with addiction potential are widely used by people of virtually all cultures in a non-addictive way. In order to understand this behaviour, its population penetrance, and its persistence, drug instrumentalization was suggested as a driving force for this consumption. Drug instrumentalization theory holds that psychoactive drugs are consumed in a very systematic way in order to make other, non-drug-related behaviours more efficient. Here, we review the evolutionary origin of this behaviour and its psychological mechanisms and explore the neurobiological and neuropharmacological mechanisms underlying them. Instrumentalization goals are discussed, for which an environmentally selective and mental state-dependent consumption of psychoactive drugs can be learned and maintained in a non-addictive way. A small percentage of people who regularly instrumentalize psychoactive drugs make a transition to addiction, which often starts with qualitative and quantitative changes in the instrumentalization goals. As such, addiction is proposed to develop from previously established long-term drug instrumentalization. Thus, preventing and treating drug addiction in an individualized medicine approach may essentially require understanding and supporting personal instrumentalization goals.
Collapse
Affiliation(s)
- Christian P Müller
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany.
| |
Collapse
|
36
|
Sagheddu C, Pintori N, Kalaba P, Dragačević V, Piras G, Lubec J, Simola N, De Luca MA, Lubec G, Pistis M. Neurophysiological and Neurochemical Effects of the Putative Cognitive Enhancer ( S)-CE-123 on Mesocorticolimbic Dopamine System. Biomolecules 2020; 10:biom10050779. [PMID: 32443397 PMCID: PMC7277835 DOI: 10.3390/biom10050779] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 12/14/2022] Open
Abstract
Treatments for cognitive impairments associated with neuropsychiatric disorders, such as attention deficit hyperactivity disorder or narcolepsy, aim at modulating extracellular dopamine levels in the brain. CE-123 (5-((benzhydrylsulfinyl)methyl) thiazole) is a novel modafinil analog with improved specificity and efficacy for dopamine transporter inhibition that improves cognitive and motivational processes in experimental animals. We studied the neuropharmacological and behavioral effects of the S-enantiomer of CE-123 ((S)-CE-123) and R-modafinil in cognitive- and reward-related brain areas of adult male rats. In vivo single unit recordings in anesthetized animals showed that (S)-CE-123, but not R-modafinil, dose-dependently (1.25 to 10 mg/kg i.v.) reduced firing of pyramidal neurons in the infralimbic/prelimbic (IL/PrL) cortex. Neither compound the affected firing activity of ventral tegmental area dopamine cells. In freely moving animals, (S)-CE-123 (10 mg/kg i.p.) increased extracellular dopamine levels in the IL/PrL, with different patterns when compared to R-modafinil (10 mg/kg i.p.); in the nucleus accumbens shell, a low and transitory increase of dopamine was observed only after (S)-CE-123. Neither (S)-CE-123 nor R-modafinil initiated the emission of 50-kHz ultrasonic vocalizations, a behavioral marker of positive affect and drug-mediated reward. Our data support previous reports of the procognitive effects of (S)-CE-123, and show a minor impact on reward-related dopaminergic areas.
Collapse
Affiliation(s)
- Claudia Sagheddu
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy; (C.S.); (N.P.); (G.P.); (N.S.); (M.A.D.L.)
| | - Nicholas Pintori
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy; (C.S.); (N.P.); (G.P.); (N.S.); (M.A.D.L.)
| | - Predrag Kalaba
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1010 Vienna, Austria; (P.K.); (V.D.)
| | - Vladimir Dragačević
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1010 Vienna, Austria; (P.K.); (V.D.)
| | - Gessica Piras
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy; (C.S.); (N.P.); (G.P.); (N.S.); (M.A.D.L.)
| | - Jana Lubec
- Department of Neuroproteomics, Paracelsus Medical University, 5020 Salzburg, Austria;
| | - Nicola Simola
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy; (C.S.); (N.P.); (G.P.); (N.S.); (M.A.D.L.)
| | - Maria Antonietta De Luca
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy; (C.S.); (N.P.); (G.P.); (N.S.); (M.A.D.L.)
| | - Gert Lubec
- Department of Neuroproteomics, Paracelsus Medical University, 5020 Salzburg, Austria;
- Correspondence: (G.L.); (M.P.); Tel.: +43-(0)-6622420-0 (G.L.); +39-070-675-4324 (M.P.)
| | - Marco Pistis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy; (C.S.); (N.P.); (G.P.); (N.S.); (M.A.D.L.)
- Neuroscience Institute, National Research Council of Italy (CNR), Section of Cagliari, 09100 Cagliari, Italy
- Correspondence: (G.L.); (M.P.); Tel.: +43-(0)-6622420-0 (G.L.); +39-070-675-4324 (M.P.)
| |
Collapse
|
37
|
Emergence of negative affect as motivation for drug taking in rats chronically self-administering cocaine. Psychopharmacology (Berl) 2020; 237:1407-1420. [PMID: 32009196 DOI: 10.1007/s00213-020-05468-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 01/21/2020] [Indexed: 12/28/2022]
Abstract
RATIONALE The role of negative affect as a motivational factor in animal models of drug addiction has been underexplored in the context of cocaine self-administration. OBJECTIVES The present investigation studied the relationship between magnitude of affective response and quantity of cocaine consumed in order to clarify the affective components that drive drug use in a preclinical model. METHODS Rats self-administered (SA) cocaine 6 h/day for 14 consecutive days while their ultrasonic vocalizations (USVs) were recorded. RESULTS Animals displayed an increase in 50-kHz call rates (indicating positive affect) when their drug levels were rapidly rising and an increase in 22-kHz call rates (indicating negative affect) when forced to abstain. The rate of 50-kHz calls predicted drug consumption during the 1st week of SA, but not week two. Contrarily, there was a strongly predictive positive association between rate of 22-kHz calls and amount of drug consumed during the 2nd week of SA. CONCLUSIONS Experimental results indicate that after chronic cocaine self-administration, negative affect emerges when animals are deprived of expected drug during withdrawal. Moreover, the increase in USVs indicating negative affect when deprived of drug was directly related to drug intake, concurrent with a decay in the direct relationship between USVs indicating positive affect and drug intake. The present preclinical support for the widely hypothesized shift from positive to negative affect as a salient motivational factor in human drug abuse adds to growing evidence of the unique value of rat USVs for understanding the role of emotion in drug addiction.
Collapse
|
38
|
Matt SM, Gaskill PJ. Where Is Dopamine and how do Immune Cells See it?: Dopamine-Mediated Immune Cell Function in Health and Disease. J Neuroimmune Pharmacol 2020; 15:114-164. [PMID: 31077015 PMCID: PMC6842680 DOI: 10.1007/s11481-019-09851-4] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/07/2019] [Indexed: 02/07/2023]
Abstract
Dopamine is well recognized as a neurotransmitter in the brain, and regulates critical functions in a variety of peripheral systems. Growing research has also shown that dopamine acts as an important regulator of immune function. Many immune cells express dopamine receptors and other dopamine related proteins, enabling them to actively respond to dopamine and suggesting that dopaminergic immunoregulation is an important part of proper immune function. A detailed understanding of the physiological concentrations of dopamine in specific regions of the human body, particularly in peripheral systems, is critical to the development of hypotheses and experiments examining the effects of physiologically relevant dopamine concentrations on immune cells. Unfortunately, the dopamine concentrations to which these immune cells would be exposed in different anatomical regions are not clear. To address this issue, this comprehensive review details the current information regarding concentrations of dopamine found in both the central nervous system and in many regions of the periphery. In addition, we discuss the immune cells present in each region, and how these could interact with dopamine in each compartment described. Finally, the review briefly addresses how changes in these dopamine concentrations could influence immune cell dysfunction in several disease states including Parkinson's disease, multiple sclerosis, rheumatoid arthritis, inflammatory bowel disease, as well as the collection of pathologies, cognitive and motor symptoms associated with HIV infection in the central nervous system, known as NeuroHIV. These data will improve our understanding of the interactions between the dopaminergic and immune systems during both homeostatic function and in disease, clarify the effects of existing dopaminergic drugs and promote the creation of new therapeutic strategies based on manipulating immune function through dopaminergic signaling. Graphical Abstract.
Collapse
Affiliation(s)
- S M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - P J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA.
| |
Collapse
|
39
|
Ahmed SH, Badiani A, Miczek KA, Müller CP. Non-pharmacological factors that determine drug use and addiction. Neurosci Biobehav Rev 2020; 110:3-27. [PMID: 30179633 PMCID: PMC6395570 DOI: 10.1016/j.neubiorev.2018.08.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/26/2018] [Accepted: 08/28/2018] [Indexed: 12/26/2022]
Abstract
Based on their pharmacological properties, psychoactive drugs are supposed to take control of the natural reward system to finally drive compulsory drug seeking and consumption. However, psychoactive drugs are not used in an arbitrary way as pure pharmacological reinforcement would suggest, but rather in a highly specific manner depending on non-pharmacological factors. While pharmacological effects of psychoactive drugs are well studied, neurobiological mechanisms of non-pharmacological factors are less well understood. Here we review the emerging neurobiological mechanisms beyond pharmacological reinforcement which determine drug effects and use frequency. Important progress was made on the understanding of how the character of an environment and social stress determine drug self-administration. This is expanded by new evidence on how behavioral alternatives and opportunities for drug instrumentalization generate different patterns of drug choice. Emerging evidence suggests that the neurobiology of non-pharmacological factors strongly determines pharmacological and behavioral drug action and may, thus, give rise for an expanded system's approach of psychoactive drug use and addiction.
Collapse
Affiliation(s)
- Serge H Ahmed
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 146 rue Léo-Saignat, F-33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 146 rue Léo-Saignat, F-33000 Bordeaux, France
| | - Aldo Badiani
- Department of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; Sussex Addiction Research and Intervention Centre (SARIC), School of Psychology, University of Sussex, BN1 9RH Brighton, UK
| | - Klaus A Miczek
- Psychology Department, Tufts University, Bacon Hall, 530 Boston Avenue, Medford, MA 02155, USA; Department of Neuroscience, Sackler School of Graduate Biomedical Sciences, Boston, MA 02111, USA
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054 Erlangen, Germany.
| |
Collapse
|
40
|
The rate of dasotraline brain entry is slow following intravenous administration. Psychopharmacology (Berl) 2020; 237:3435-3446. [PMID: 32813030 PMCID: PMC7651685 DOI: 10.1007/s00213-020-05623-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 07/27/2020] [Indexed: 12/03/2022]
Abstract
RATIONALE Drugs that rapidly increase dopamine levels have an increased risk of abuse. Dasotraline (DAS) is a dopamine and norepinephrine reuptake inhibitor characterized by slow oral absorption with low potential for abuse. However, it remains unclear whether intravenous (i.v.) administration would facilitate the rapid elevation of dopamine levels associated with stimulant drugs. OBJECTIVE To assess the kinetics of DAS across the blood-brain barrier and time to onset of dopamine transporters (DAT) inhibition. METHODS We compared the onset of DAT occupancy and the associated elevation of synaptic dopamine levels in rhesus monkey following i.v. administration of DAS or methylphenidate (MPH) using positron emission tomography (PET). Brain entry times were estimated by reductions in [18F]-FE-PE2I binding to DAT in rhesus monkeys. Elevations of synaptic dopamine were estimated by reductions in [11C]-Raclopride binding to D2 receptors. RESULTS Intravenous administration of DAS (0.1 and 0.2 mg/kg) resulted in striatal DAT occupancies of 54% and 68%, respectively; i.v. administered MPH (0.1 and 0.5 mg/kg) achieved occupancies of 69% and 88% respectively. Brain entry times of DAS (22 and 15 min, respectively) were longer than for MPH (3 and 2 min). Elevations in synaptic dopamine were similar for both DAS and MPH however the time for half-maximal displacement by MPH (t = 23 min) was 4-fold more rapid than for DAS (t = 88 min). CONCLUSIONS These results demonstrate that the pharmacodynamics effects of DAS on DAT occupancy and synaptic dopamine levels are more gradual in onset than those of MPH even with i.v. administration that is favored by recreational drug abusers.
Collapse
|
41
|
Lack of effect of the combination of metyrapone and oxazepam on brain dopamine. Brain Res 2019; 1724:146435. [DOI: 10.1016/j.brainres.2019.146435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/20/2019] [Accepted: 09/02/2019] [Indexed: 01/04/2023]
|
42
|
Fu Y, Depue RA. A novel neurobehavioral framework of the effects of positive early postnatal experience on incentive and consummatory reward sensitivity. Neurosci Biobehav Rev 2019; 107:615-640. [DOI: 10.1016/j.neubiorev.2019.09.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 09/08/2019] [Accepted: 09/17/2019] [Indexed: 12/22/2022]
|
43
|
Tanabe J, Regner M, Sakai J, Martinez D, Gowin J. Neuroimaging reward, craving, learning, and cognitive control in substance use disorders: review and implications for treatment. Br J Radiol 2019; 92:20180942. [PMID: 30855982 PMCID: PMC6732921 DOI: 10.1259/bjr.20180942] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/13/2019] [Accepted: 02/21/2019] [Indexed: 01/17/2023] Open
Abstract
Substance use disorder is a leading causes of preventable disease and mortality. Drugs of abuse cause molecular and cellular changes in specific brain regions and these neuroplastic changes are thought to play a role in the transition to uncontrolled drug use. Neuroimaging has identified neural substrates associated with problematic substance use and may offer clues to reduce its burden on the patient and society. Here, we provide a narrative review of neuroimaging studies that have examined the structures and circuits associated with reward, cues and craving, learning, and cognitive control in substance use disorders. Most studies use advanced MRI or positron emission tomography (PET). Many studies have focused on the dopamine neurons of the ventral tegmental area, and the regions where these neurons terminate, such as the striatum and prefrontal cortex. Decreases in dopamine receptors and transmission have been found in chronic users of drugs, alcohol, and nicotine. Recent studies also show evidence of differences in structure and function in substance users relative to controls in brain regions involved in salience evaluation, such as the insula and anterior cingulate cortex. Balancing between reward-related bottom-up and cognitive-control-related top-down processes is discussed in the context of neuromodulation as a potential treatment. Finally, some of the challenges for understanding substance use disorder using neuroimaging methods are discussed.
Collapse
Affiliation(s)
| | - Michael Regner
- Department of Radiology, University of Colorado Anschutz Medical Center, Aurora, CO
| | - Joseph Sakai
- Department of Psychiatry, University of Colorado Anschutz Medical Center, Aurora, CO
| | - Diana Martinez
- Department of Psychiatry, Columbia University, New York, USA
| | | |
Collapse
|
44
|
Abstract
The human dopamine transporter gene SLC6A3 is involved in substance use disorders (SUDs) among many other common neuropsychiatric illnesses but allelic association results including those with its classic genetic markers 3'VNTR or Int8VNTR remain mixed and unexplainable. To better understand the genetics for reproducible association signals, we report the presence of recombination hotspots based on sequencing of the entire 5' promoter regions in two small SUDs cohorts, 30 African Americans (AAs) and 30 European Americans (EAs). Recombination rate was the highest near the transcription start site (TSS) in both cohorts. In addition, each cohort carried 57 different promoter haplotypes out of 60 and no haplotypes were shared between the two ethnicities. A quarter of the haplotypes evolved in an ethnicity-specific manner. Finally, analysis of five hundred subjects of European ancestry, from the 1000 Genome Project, confirmed the promoter recombination hotspots and also revealed several additional ones in non-coding regions only. These findings provide an explanation for the mixed results as well as guidance for selection of effective markers to be used in next generation association validation (NGAV), facilitating the delineation of pathogenic variation in this critical neuropsychiatric gene.
Collapse
|
45
|
Pei F, Li H, Liu B, Bahar I. Quantitative Systems Pharmacological Analysis of Drugs of Abuse Reveals the Pleiotropy of Their Targets and the Effector Role of mTORC1. Front Pharmacol 2019; 10:191. [PMID: 30906261 PMCID: PMC6418047 DOI: 10.3389/fphar.2019.00191] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/14/2019] [Indexed: 12/14/2022] Open
Abstract
Existing treatments against drug addiction are often ineffective due to the complexity of the networks of protein-drug and protein-protein interactions (PPIs) that mediate the development of drug addiction and related neurobiological disorders. There is an urgent need for understanding the molecular mechanisms that underlie drug addiction toward designing novel preventive or therapeutic strategies. The rapidly accumulating data on addictive drugs and their targets as well as advances in machine learning methods and computing technology now present an opportunity to systematically mine existing data and draw inferences on potential new strategies. To this aim, we carried out a comprehensive analysis of cellular pathways implicated in a diverse set of 50 drugs of abuse using quantitative systems pharmacology methods. The analysis of the drug/ligand-target interactions compiled in DrugBank and STITCH databases revealed 142 known and 48 newly predicted targets, which have been further analyzed to identify the KEGG pathways enriched at different stages of drug addiction cycle, as well as those implicated in cell signaling and regulation events associated with drug abuse. Apart from synaptic neurotransmission pathways detected as upstream signaling modules that “sense” the early effects of drugs of abuse, pathways involved in neuroplasticity are distinguished as determinants of neuronal morphological changes. Notably, many signaling pathways converge on important targets such as mTORC1. The latter emerges as a universal effector of the persistent restructuring of neurons in response to continued use of drugs of abuse.
Collapse
Affiliation(s)
- Fen Pei
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Hongchun Li
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Bing Liu
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ivet Bahar
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
46
|
Abstract
Numerous drugs elicit locomotor stimulant effects at appropriate doses; however, we typically reserve the term psychostimulant to refer to drugs with affinity for monoamine reuptake transporters. This chapter comprises select experiments that have characterized the discriminative stimulus effects of psychostimulants using drug discrimination procedures. The substitution profiles of psychostimulants in laboratory rodents are generally consistent with those observed in human and nonhuman primate drug discrimination experiments. Notably, two major classes of psychostimulants can be distinguished as those that function as passive monoamine reuptake inhibitors (such as cocaine) and those that function as substrates for monoamine transporters and stimulate monoamine release (such as the amphetamines). Nevertheless, the discriminative stimulus effects of both classes of psychostimulant are quite similar, and drugs from different classes will substitute for one another. Most importantly, for both the cocaine-like and amphetamine-like psychostimulants, dopaminergic mechanisms most saliently determine discriminative stimulus effects, but these effects can be modulated by alterations in noradrenergic and serotonergic neurotransmission as well. Thusly, the drug discrimination assay is useful for characterizing the interoceptive effects of psychostimulants and determining the mechanisms that contribute to their subjective effects in humans.
Collapse
Affiliation(s)
- Michael D Berquist
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 West Markham Street, Slot 638, Little Rock, AR, 72205, USA
| | - William E Fantegrossi
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 West Markham Street, Slot 638, Little Rock, AR, 72205, USA.
| |
Collapse
|
47
|
López AJ, Siciliano CA, Calipari ES. Activity-Dependent Epigenetic Remodeling in Cocaine Use Disorder. Handb Exp Pharmacol 2019; 258:231-263. [PMID: 31628597 DOI: 10.1007/164_2019_257] [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: 12/27/2022]
Abstract
Substance use disorder (SUD) is a behavioral disorder characterized by cycles of abstinence, drug seeking, and relapse. SUD is characterized by aberrant learning processes which develop after repeated exposure to drugs of abuse. At the core of this phenotype is the persistence of symptoms, such as craving and relapse to drug seeking, long after the cessation of drug use. The neural basis of these behavioral changes has been linked to dysfunction in neural circuits across the brain; however, the molecular drivers that allow for these changes to persist beyond the lifespan of any individual protein remain opaque. Epigenetic adaptations - where DNA is modified to increase or decrease the probability of gene expression at key genes - have been identified as a mechanism underlying the long-lasting nature of drug-seeking behavior. Thus, to understand SUD, it is critical to define the interplay between neuronal activation and longer-term changes in transcription and epigenetic remodeling and define their role in addictive behaviors. In this review, we discuss the current understanding of drug-induced changes to circuit function, recent discoveries in epigenetic mechanisms that mediate these changes, and, ultimately, how these adaptations drive the persistent nature of relapse, with emphasis on adaptations in models of cocaine use disorder. Understanding the complex interplay between epigenetic gene regulation and circuit activity will be critical in elucidating the neural mechanisms underlying SUD. This, with the advent of novel genetic-based techniques, will allow for the generation of novel therapeutic avenues to improve treatment outcomes in SUD.
Collapse
Affiliation(s)
- Alberto J López
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Erin S Calipari
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA. .,Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN, USA. .,Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA. .,Department of Molecular Physiology and Biophysics, Vanderbilt Institute for Infection, Immunology, and Infection, Vanderbilt University School of Medicine, Nashville, TN, USA. .,Department of Psychiatry and Behavioral Sciences, Vanderbilt Institute for Infection, Immunology, and Infection, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
48
|
Amato D, Kruyer A, Samaha AN, Heinz A. Hypofunctional Dopamine Uptake and Antipsychotic Treatment-Resistant Schizophrenia. Front Psychiatry 2019; 10:314. [PMID: 31214054 PMCID: PMC6557273 DOI: 10.3389/fpsyt.2019.00314] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/23/2019] [Indexed: 01/07/2023] Open
Abstract
Antipsychotic treatment resistance in schizophrenia remains a major issue in psychiatry. Nearly 30% of patients with schizophrenia do not respond to antipsychotic treatment, yet the underlying neurobiological causes are unknown. All effective antipsychotic medications are thought to achieve their efficacy by targeting the dopaminergic system. Here we review early literature describing the fundamental mechanisms of antipsychotic drug efficacy, highlighting mechanistic concepts that have persisted over time. We then reconsider the original framework for understanding antipsychotic efficacy in light of recent advances in our scientific understanding of the dopaminergic effects of antipsychotics. Based on these new insights, we describe a role for the dopamine transporter in the genesis of both antipsychotic therapeutic response and primary resistance. We believe that this discussion will help delineate the dopaminergic nature of antipsychotic treatment-resistant schizophrenia.
Collapse
Affiliation(s)
- Davide Amato
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Anna Kruyer
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Anne-Noël Samaha
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Andreas Heinz
- Department of Psychiatry, Charité University Medicine Berlin, Campus Charité Mitte, Berlin, Germany
| |
Collapse
|
49
|
Mukhara D, Banks ML, Neigh GN. Stress as a Risk Factor for Substance Use Disorders: A Mini-Review of Molecular Mediators. Front Behav Neurosci 2018; 12:309. [PMID: 30622460 PMCID: PMC6308626 DOI: 10.3389/fnbeh.2018.00309] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 11/28/2018] [Indexed: 12/15/2022] Open
Abstract
The extant literature supports the role of stress in enhancing the susceptibility of drug abuse progressing to a substance use disorder diagnosis. However, the molecular mediators by which stress enhances the progression from cocaine abuse to cocaine use disorder via the mesolimbic pathway remain elusive. In this mini-review article, we highlight three mechanisms by which glucocorticoids (GCs) and the dopaminergic system interact. First, GCs upregulate tyrosine hydroxylase (TH), the rate-limiting enzyme in dopamine (DA) synthesis. Second, GCs downregulate monoamine-oxidase (MAO), an enzyme responsible for DA removal. Lastly, GCs are hypothesized to decrease DA reuptake, subsequently increasing synaptic DA. Based on these interactions, we review preclinical literature highlighting how stress modulates the mesolimbic pathway, including the ventral tegmental area (VTA) and nucleus accumbens (NAcs), to alter cocaine abuse-related effects. Taken together, stress enhances cocaine's abuse-related effects at multiple points along the VTA mesolimbic projection, and uniquely in the NAcs through a positive feedback type mechanism. Furthermore, we highlight future directions to elucidate the interaction between the prefrontal cortex (PFC) and key intermediaries including ΔFosB, cAMP response element binding protein (CREB) and cyclin-dependent kinase 5 (CDK5) to highlight possible mechanisms that underlie stress-induced acceleration of the progression to a cocaine use disorder diagnosis.
Collapse
Affiliation(s)
- Deepika Mukhara
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA, United States
| | - Matthew L. Banks
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | - Gretchen N. Neigh
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
50
|
Carias E, Hamilton J, Robison LS, Delis F, Eiden R, Quattrin T, Hadjiargyrou M, Komatsu D, Thanos PK. Chronic oral methylphenidate treatment increases microglial activation in rats. J Neural Transm (Vienna) 2018; 125:1867-1875. [PMID: 30238340 DOI: 10.1007/s00702-018-1931-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/17/2018] [Indexed: 12/18/2022]
Abstract
Methylphenidate (MP) is a widely prescribed psychostimulant used to treat attention deficit hyperactivity disorder. Previously, we established a drinking paradigm to deliver MP to rats at doses that result in pharmacokinetic profiles similar to treated patients. In the present study, adolescent male rats were assigned to one of three groups: control (water), low-dose MP (LD; 4/10 mg/kg), and high dose MP (HD; 30/60 mg/kg). Following 3 months of treatment, half of the rats in each group were euthanized, and the remaining rats received only water throughout a 1-month-long abstinence phase. In vitro autoradiography using [3H] PK 11195 was performed to measure microglial activation. HD MP rats showed increased [3H] PK 11195 binding compared to control rats in several cerebral cortical areas: primary somatosensory cortex including jaw (68.6%), upper lip (80.1%), barrel field (88.9%), and trunk (78%) regions, forelimb sensorimotor area (87.3%), secondary somatosensory cortex (72.5%), motor cortices 1 (73.2%) and 2 (69.3%), insular cortex (59.9%); as well as subcortical regions including the thalamus (62.9%), globus pallidus (79.4%) and substantia nigra (22.7%). Additionally, HD MP rats showed greater binding compared to LD MP rats in the hippocampus (60.6%), thalamus (59.6%), substantia nigra (38.5%), and motor 2 cortex (55.3%). Following abstinence, HD MP rats showed no significant differences compared to water controls; however, LD MP rats showed increased binding in pre-limbic cortex (78.1%) and ventromedial caudate putamen (113.8%). These findings indicate that chronic MP results in widespread microglial activation immediately after treatment and following the cessation of treatment in some brain regions.
Collapse
Affiliation(s)
- Emily Carias
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 1021 Main St., Buffalo, NY, USA
| | - John Hamilton
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 1021 Main St., Buffalo, NY, USA
| | - Lisa S Robison
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave., Albany, NY, USA
| | - Foteini Delis
- Department of Pharmacology, Medical School, University of Ioannina, 45110, Ioannina, Greece
| | - Rina Eiden
- Department of Psychology, University at Buffalo, 1021 Main St., Buffalo, NY, USA
| | - Teresa Quattrin
- Women and Children's Hospital of Buffalo, Department of Pediatrics, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Michael Hadjiargyrou
- Department of Life Sciences, New York Institute of Technology, Northern Blvd., Old Westbury, NY, USA
| | - David Komatsu
- Department of Orthopedics, Stony Brook University, 100 Nicolls Rd., Stony Brook, NY, USA
| | - Panayotis K Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 1021 Main St., Buffalo, NY, USA.
| |
Collapse
|