1
|
CDH2 mutation affecting N-cadherin function causes attention-deficit hyperactivity disorder in humans and mice. Nat Commun 2021; 12:6187. [PMID: 34702855 PMCID: PMC8548587 DOI: 10.1038/s41467-021-26426-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/29/2021] [Indexed: 11/20/2022] Open
Abstract
Attention-deficit hyperactivity disorder (ADHD) is a common childhood-onset psychiatric disorder characterized by inattention, impulsivity and hyperactivity. ADHD exhibits substantial heritability, with rare monogenic variants contributing to its pathogenesis. Here we demonstrate familial ADHD caused by a missense mutation in CDH2, which encodes the adhesion protein N-cadherin, known to play a significant role in synaptogenesis; the mutation affects maturation of the protein. In line with the human phenotype, CRISPR/Cas9-mutated knock-in mice harboring the human mutation in the mouse ortholog recapitulated core behavioral features of hyperactivity. Symptoms were modified by methylphenidate, the most commonly prescribed therapeutic for ADHD. The mutated mice exhibited impaired presynaptic vesicle clustering, attenuated evoked transmitter release and decreased spontaneous release. Specific downstream molecular pathways were affected in both the ventral midbrain and prefrontal cortex, with reduced tyrosine hydroxylase expression and dopamine levels. We thus delineate roles for CDH2-related pathways in the pathophysiology of ADHD. Molecular mechanisms of attention-deficit hyperactivity disorder (ADHD) are not fully understood. Here the authors demonstrate a mutation in CDH2, encoding N-cadherin, that is associated with ADHD, and in a mouse model, delineate molecular electrophysiological characteristics associated with this mutation.
Collapse
|
2
|
Olsen D, Wellner N, Kaas M, de Jong IEM, Sotty F, Didriksen M, Glerup S, Nykjaer A. Altered dopaminergic firing pattern and novelty response underlie ADHD-like behavior of SorCS2-deficient mice. Transl Psychiatry 2021; 11:74. [PMID: 33495438 PMCID: PMC7835366 DOI: 10.1038/s41398-021-01199-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 12/12/2020] [Accepted: 01/05/2021] [Indexed: 01/03/2023] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is the most frequently diagnosed neurodevelopmental disorder worldwide. Affected individuals present with hyperactivity, inattention, and cognitive deficits and display a characteristic paradoxical response to drugs affecting the dopaminergic system. However, the underlying pathophysiology of ADHD and how this relates to dopaminergic transmission remains to be fully understood. Sorcs2-/- mice uniquely recapitulate symptoms reminiscent of ADHD in humans. Here, we show that lack of SorCS2 in mice results in lower sucrose intake, indicating general reward deficits. Using in-vivo recordings, we further find that dopaminergic transmission in the ventral tegmental area (VTA) is shifted towards a more regular firing pattern with marked reductions in the relative occurrence of irregular firing in Sorcs2-/- mice. This was paralleled by abnormal acute behavioral responses to dopamine receptor agonists, suggesting fundamental differences in dopaminergic circuits and indicating a perturbation in the balance between the activities of the postsynaptic dopamine receptor DRD1 and the presynaptic inhibitory autoreceptor DRD2. Interestingly, the hyperactivity and drug response of Sorcs2-/- mice were markedly affected by novelty. Taken together, our findings show how loss of a candidate ADHD-risk gene has marked effects on dopaminergic circuit function and the behavioral response to the environment.
Collapse
Affiliation(s)
- Ditte Olsen
- grid.7048.b0000 0001 1956 2722Department of Biomedicine, Aarhus University, Hoegh-Guldbergsgade 10, DK-8000 Aarhus C, Denmark ,grid.424580.f0000 0004 0476 7612Neurodegeneration and Biologics, H. Lundbeck A/S, Ottiliavej 9, DK-2500 Valby, Denmark ,grid.7048.b0000 0001 1956 2722Present Address: Department of Biomedicine, Aarhus University, Hoegh-Guldbergsgade 10, DK-8000 Aarhus C, Denmark
| | - Niels Wellner
- grid.7048.b0000 0001 1956 2722Department of Biomedicine, Aarhus University, Hoegh-Guldbergsgade 10, DK-8000 Aarhus C, Denmark ,grid.7048.b0000 0001 1956 2722Danish Research Institute of Translational Neuroscience DANDRITE Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Hoegh-Guldbergsgade 10, DK-8000 Aarhus C, Denmark
| | - Mathias Kaas
- grid.7048.b0000 0001 1956 2722Department of Biomedicine, Aarhus University, Hoegh-Guldbergsgade 10, DK-8000 Aarhus C, Denmark
| | - Inge E. M. de Jong
- grid.424580.f0000 0004 0476 7612Neurodegeneration and Biologics, H. Lundbeck A/S, Ottiliavej 9, DK-2500 Valby, Denmark
| | - Florence Sotty
- grid.424580.f0000 0004 0476 7612Neurodegeneration and Biologics, H. Lundbeck A/S, Ottiliavej 9, DK-2500 Valby, Denmark
| | - Michael Didriksen
- grid.424580.f0000 0004 0476 7612Neurodegeneration and Biologics, H. Lundbeck A/S, Ottiliavej 9, DK-2500 Valby, Denmark
| | - Simon Glerup
- Department of Biomedicine, Aarhus University, Hoegh-Guldbergsgade 10, DK-8000, Aarhus C, Denmark.
| | - Anders Nykjaer
- Department of Biomedicine, Aarhus University, Hoegh-Guldbergsgade 10, DK-8000, Aarhus C, Denmark. .,Danish Research Institute of Translational Neuroscience DANDRITE Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Hoegh-Guldbergsgade 10, DK-8000, Aarhus C, Denmark. .,The Danish National Research Foundation Center PROMEMO, Aarhus University, Hoegh-Guldbergsgade 10, DK-8000, Aarhus C, Denmark. .,Department of Neurosurgery, Skejby University Hospital, Palle Juul-Jensens Blvd. 99, DK-8200, Aarhus N, Denmark.
| |
Collapse
|
3
|
Schmill MP, Cadney MD, Thompson Z, Hiramatsu L, Albuquerque RL, McNamara MP, Castro AA, Kay JC, Buenaventura DG, Ramirez JL, Rhodes JS, Garland T. Conditioned place preference for cocaine and methylphenidate in female mice from lines selectively bred for high voluntary wheel-running behavior. GENES BRAIN AND BEHAVIOR 2020; 20:e12700. [PMID: 32909333 DOI: 10.1111/gbb.12700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 12/27/2022]
Abstract
Behavioral addictions can come in many forms, including overeating, gambling and overexercising. All addictions share a common mechanism involving activation of the natural reward circuit and reinforcement learning, but the extent to which motivation for natural and drug rewards share similar neurogenetic mechanisms remains unknown. A unique mouse genetic model in which four replicate lines of female mice were selectively bred (>76 generations) for high voluntary wheel running (High Runner or HR lines) alongside four non-selected control (C) lines were used to test the hypothesis that high motivation for exercise is associated with greater reward for cocaine (20 mg/kg) and methylphenidate (10 mg/kg) using the conditioned place preference (CPP) test. HR mice run ~three times as many revolutions/day as C mice, but the extent to which they have increased motivation for other rewards is unknown. Both HR and C mice displayed significant CPP for cocaine and methylphenidate, but with no statistical difference between linetypes for either drug. Taken together, results suggest that selective breeding for increased voluntary running has modified the reward circuit in the brain in a way that increases motivation for running without affecting cocaine or methylphenidate reward.
Collapse
Affiliation(s)
- Margaret P Schmill
- Neuroscience Graduate Program, University of California, Riverside, California, USA
| | - Marcell D Cadney
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, California, USA
| | - Zoe Thompson
- Neuroscience Graduate Program, University of California, Riverside, California, USA
| | - Layla Hiramatsu
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, California, USA
| | - Ralph L Albuquerque
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, California, USA
| | - Monica P McNamara
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, California, USA
| | - Alberto A Castro
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, California, USA
| | - Jarren C Kay
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, California, USA
| | - Darrius G Buenaventura
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, California, USA
| | - Jocelyn L Ramirez
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, California, USA
| | - Justin S Rhodes
- Beckman Institute for Advanced Science and Technology, Urbana, Illinois, USA.,Center for Nutrition, Learning and Memory, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Theodore Garland
- Neuroscience Graduate Program, University of California, Riverside, California, USA.,Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, California, USA
| |
Collapse
|
4
|
Joshi K, Chandra A, Jain K, Talegaonkar S. Nanocrystalization: An Emerging Technology to Enhance the Bioavailability of Poorly Soluble Drugs. Pharm Nanotechnol 2019; 7:259-278. [PMID: 30961518 PMCID: PMC6967137 DOI: 10.2174/2211738507666190405182524] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/18/2019] [Accepted: 04/01/2019] [Indexed: 01/08/2023]
Abstract
Most of the active pharmaceutical ingredient used in the management of disease have poor water solubility and offer grueling problems in drug formulation development since low solubility is generally associated with poor dissolution characteristics which leads to poor oral bioavailability. The great challenge for the development of a pharmaceutical product is to create its new formulation and drug delivery system to limit solubility problems of existing drug candidate. Limited drug-loading capacity requires a large amount of carrier material to get appropriate encapsulation of the drug, which is another major challenge in the development of pharmaceutical product which could be resolved by developing nanocrystals (NCs). A significant research in the past few years has been done to develop NCs which helps in the delivery of poorly water soluble drugs via different routes. The technology could continue to thrive as a useful tool in pharmaceutical sciences for the improvement of drug solubility, absorption and bioavailability. Many crystalline compounds have pulled in incredible consideration much of the time, due to their ability to show good physical and chemical properties when contrasted with their amorphous counterparts. Nanocrystals have been proven to show atypical properties compared to the bulk. This review article explores the principles of the important nanocrystallization techniques including NCs characterization and its application.
Collapse
Affiliation(s)
| | | | | | - Sushama Talegaonkar
- Address correspondence to this author at the Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar Sec III, New Delhi-110017, India and Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-62, India; Tel: 9818453518; E-mail:
| |
Collapse
|
5
|
Jager A, Kanters D, Geers F, Buitelaar JK, Kozicz T, Glennon JC. Methylphenidate Dose-Dependently Affects Aggression and Improves Fear Extinction and Anxiety in BALB/cJ Mice. Front Psychiatry 2019; 10:768. [PMID: 31708820 PMCID: PMC6823535 DOI: 10.3389/fpsyt.2019.00768] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/24/2019] [Indexed: 12/22/2022] Open
Abstract
Overt aggression, increased anxiety, and dysfunctional fear processing are often observed in individuals with conduct disorder (CD) and attention-deficit hyperactivity disorder (ADHD). Methylphenidate (MPH), a psychostimulant increasing dopamine and noradrenaline tone, is effective in reducing aggression in both CD and ADHD individuals. However, it is unclear to which extent these effects of MPH are dose dependent. Here, the effects of acute intraperitoneal MPH (3 and 10 mg/kg) on aggression, anxiety, social behavior, and fear extinction were investigated in BALB/cJ mice. Previous studies in BALB/cJ mice have revealed high levels of aggression and anxiety that are associated with reduced top-down cortical control. Administration of 3 mg/kg MPH prolonged the attack latency and prevented escalation of aggression over time compared to vehicle-treated mice, while 10 mg/kg MPH increased number of bites and attacks. In addition, 3 mg/kg MPH decreased social interaction slightly. A strong anxiolytic effect was found after administration of both the 3 and 10 mg/kg doses in the elevated plus maze and the open-field test. In addition, while vehicle-treated BALB/cJ animals showed intact freezing, both doses of MPH decreased freezing to the unconditioned stimulus in a fear-conditioning paradigm. A long-lasting effect on fear extinction was visible after treatment with the 10 mg/kg dose. The data support a role for MPH in the regulation of anxiety, fear processing, and aggression in BALB/cJ mice, with the latter effect in a dose-dependent manner. The findings provide a further context for examining the effects of MPH in clinical disorders such as ADHD and CD.
Collapse
Affiliation(s)
- Amanda Jager
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboudumc, Nijmegen, Netherlands
| | - Doranda Kanters
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboudumc, Nijmegen, Netherlands
| | - Femke Geers
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboudumc, Nijmegen, Netherlands
| | - Jan K Buitelaar
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboudumc, Nijmegen, Netherlands
| | - Tamas Kozicz
- Department of Anatomy, Donders Institute for Brain, Cognition and Behavior, Radboudumc, Nijmegen, Netherlands.,Department of Clinical Genomics, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States
| | - Jeffrey C Glennon
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboudumc, Nijmegen, Netherlands
| |
Collapse
|
6
|
Foroughi K, Khaksari M, Shayannia A. Molecular Docking Studies of Methamphetamine and Amphetamine- Related Derivatives as an Inhibitor against Dopamine Receptor. Curr Comput Aided Drug Des 2018; 16:122-133. [PMID: 30514192 DOI: 10.2174/1573409915666181204144411] [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: 09/17/2018] [Revised: 11/14/2018] [Accepted: 11/29/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND The catecholamines such as dopamine, norepinephrine, and epinephrine are neurotransmitters that regulate different physiological functions of the central nervous system. Some evidence suggests that the degeneration of dopamine neurons in the substantia nigra contributes to Parkinson's Disease (PD), which is a neurodegenerative disorder and it is responsible for the major symptoms of PD. It is suggested that replenishment of striatal dopamine through the oral administration of the dopamine precursor, levodopa, can compensate for the lack of endogenously produced dopamine. Some studies have shown competitive inhibition of dopamine receptor such as methamphetamine, and other amphetamine-related derivatives, which block dopamine receptor activity to uptake dopamine. METHODS In this study, 3D structures of amphetamine, methamphetamine, cocaine, methylphenidate, cathinone, MDMA, and mephedrone were obtained from the PubChem database, which has reported some evidence about their inhibitory effect with dopamine receptor. Then, these structures were provided for molecular docking analysis by Autodock Vina software. Eventually, the binding energies between docked dopamine receptor and them were calculated and their interactions were prognosticated. RESULTS Our results indicated that all chemicals can interact with dopamine receptor molecule in the active site of dopamine and the minimum binding energies belong to Cocaine and Methylphenidate with -7.9 Kcal/mol and -7.2 Kcal/mol, respectively. CONCLUSION It might be concluded that amphetamine, methamphetamine, cocaine, methylphenidate, cathinone, MDMA, and mephedrone could act as potential inhibitors of DA receptor for dopamine uptake, which could cause degenerative disorders.
Collapse
Affiliation(s)
- Kobra Foroughi
- Department of Medical Biotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mehdi Khaksari
- Addiction Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Asghar Shayannia
- Department of Medical Biotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
7
|
Wiers CE, Shokri-Kojori E, Wong CT, Abi-Dargham A, Demiral ŞB, Tomasi D, Wang GJ, Volkow ND. Cannabis Abusers Show Hypofrontality and Blunted Brain Responses to a Stimulant Challenge in Females but not in Males. Neuropsychopharmacology 2016; 41:2596-605. [PMID: 27156854 PMCID: PMC4987858 DOI: 10.1038/npp.2016.67] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 04/27/2016] [Accepted: 04/30/2016] [Indexed: 12/14/2022]
Abstract
The extent to which cannabis is deleterious to the human brain is not well understood. Here, we test whether cannabis abusers (CA) have impaired frontal function and reactivity to dopaminergic signaling, which are fundamental to relapse in addiction. We measured brain glucose metabolism using PET and [(18)F]FDG both at baseline (placebo) and after challenge with methylphenidate (MP), a dopamine-enhancing drug, in 24 active CA (50% female) and 24 controls (HC; 50% female). Results show that (i) CA had lower baseline glucose metabolism than HC in frontal cortex including anterior cingulate, which was associated with negative emotionality. (ii) MP increased whole-brain glucose metabolism in HC but not in CA; and group by challenge effects were most profound in putamen, caudate, midbrain, thalamus, and cerebellum. In CA, MP-induced metabolic increases in putamen correlated negatively with addiction severity. (iii) There were significant gender effects, such that both the group differences at baseline in frontal metabolism and the attenuated regional brain metabolic responses to MP were observed in female CA but not in male CA. As for other drug addictions, reduced baseline frontal metabolism is likely to contribute to relapse in CA. The attenuated responses to MP in midbrain and striatum are consistent with decreased brain reactivity to dopamine stimulation and might contribute to addictive behaviors in CA. The gender differences suggest that females are more sensitive than males to the adverse effects of cannabis in brain.
Collapse
Affiliation(s)
- Corinde E Wiers
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA,National Institute on Alcohol Abuse and Alcoholism, Laboratory of Neuroimaging, National Institutes of Health, 10 Center Drive, Room B2L124, Bethesda, MD 20892, USA, Tel: +1 301 451 3021 or +1 301 402 0868, Fax: +1 301 496 5568, E-mail: or
| | - Ehsan Shokri-Kojori
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Christopher T Wong
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Anissa Abi-Dargham
- Division of Translational Imaging, Department of Psychiatry, Columbia University and New York State Psychiatric Institute, New York, NY, USA
| | - Şükrü B Demiral
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Dardo Tomasi
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Gene-Jack Wang
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA,National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA,National Institute on Alcohol Abuse and Alcoholism, Laboratory of Neuroimaging, National Institutes of Health, 10 Center Drive, Room B2L124, Bethesda, MD 20892, USA, Tel: +1 301 451 3021 or +1 301 402 0868, Fax: +1 301 496 5568, E-mail: or
| |
Collapse
|
8
|
Kung TS, Richardson JR, Cooper KR, White LA. Developmental Deltamethrin Exposure Causes Persistent Changes in Dopaminergic Gene Expression, Neurochemistry, and Locomotor Activity in Zebrafish. Toxicol Sci 2015; 146:235-43. [PMID: 25912032 DOI: 10.1093/toxsci/kfv087] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Pyrethroids are commonly used insecticides that are considered to pose little risk to human health. However, there is an increasing concern that children are more susceptible to the adverse effects of pesticides. We used the zebrafish model to test the hypothesis that developmental exposure to low doses of the pyrethroid deltamethrin results in persistent alterations in dopaminergic gene expression, neurochemistry, and locomotor activity. Zebrafish embryos were treated with deltamethrin (0.25-0.50 μg/l), at concentrations below the LOAEL, during the embryonic period [3-72 h postfertilization (hpf)], after which transferred to fresh water until the larval stage (2-weeks postfertilization). Deltamethrin exposure resulted in decreased transcript levels of the D1 dopamine (DA) receptor (drd1) and increased levels of tyrosine hydroxylase at 72 hpf. The reduction in drd1 transcripts persisted to the larval stage and was associated with decreased D2 dopamine receptor transcripts. Larval fish, exposed developmentally to deltamethrin, had increased levels of homovanillic acid, a DA metabolite. Since the DA system is involved in locomotor activity, we measured the swim activity of larval fish following a transition to darkness. Developmental exposure to deltamethrin significantly increased larval swim activity which was attenuated by concomitant knockdown of the DA transporter. Acute exposure to methylphenidate, a DA transporter inhibitor, increased swim activity in control larva, while reducing swim activity in larva developmentally exposed to deltamethrin. Developmental exposure to deltamethrin causes locomotor deficits in larval zebrafish, which is likely mediated by dopaminergic dysfunction. This highlights the need to understand the persistent effects of low-dose neurotoxicant exposure during development.
Collapse
Affiliation(s)
- Tiffany S Kung
- *Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA, Department of Biochemistry and Microbiology, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA and
| | - Jason R Richardson
- Department of Environmental and Occupational Medicine, Environmental and Occupational Health Sciences Institute, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Keith R Cooper
- Department of Biochemistry and Microbiology, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA and Department of Environmental and Occupational Medicine, Environmental and Occupational Health Sciences Institute, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Lori A White
- Department of Biochemistry and Microbiology, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA and
| |
Collapse
|
9
|
Calipari ES, Ferris MJ, Siciliano CA, Jones SR. Differential influence of dopamine transport rate on the potencies of cocaine, amphetamine, and methylphenidate. ACS Chem Neurosci 2015; 6:155-62. [PMID: 25474655 PMCID: PMC4304485 DOI: 10.1021/cn500262x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
![]()
Dopamine
transporter (DAT) levels vary across brain regions and
individuals, and are altered by drug history and disease states; however,
the impact of altered DAT expression on psychostimulant effects in
brain has not been systematically explored. Using fast scan cyclic
voltammetry, we measured the effects of elevated DAT levels on presynaptic
dopamine parameters as well as the uptake inhibition potency of the
blockers cocaine and methylphenidate (MPH) and the releaser amphetamine
(AMPH) in the nucleus accumbens core. Here we found that increases
in DAT levels, resulting from either genetic overexpression or MPH
self-administration, caused markedly increased maximal rates of uptake
(Vmax) that were positively correlated
with the uptake inhibition potency of AMPH and MPH, but not cocaine.
AMPH and MPH were particularly sensitive to DAT changes, with a 100%
increase in Vmax resulting in a 200% increase
in potency. The relationship between Vmax and MPH potency was the same as that for AMPH, but was different
from that for cocaine, indicating that MPH more closely resembles
a releaser with regard to uptake inhibition. Conversely, the effects
of MPH on stimulated dopamine release were similar to those of cocaine,
with inverted U-shaped increases in release over a concentration–response
curve. This was strikingly different from the release profile of AMPH,
which showed only reductions at high concentrations, indicating that
MPH is not a pure releaser. These data indicate that although MPH
is a DAT blocker, its uptake-inhibitory actions are affected by DAT
changes in a similar manner to releasers. Together, these data show
that fluctuations in DAT levels alter the potency of releasers and
MPH but not blockers and suggest an integral role of the DAT in the
addictive potential of AMPH and related compounds.
Collapse
Affiliation(s)
- Erin S. Calipari
- Department
of Physiology
and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Mark J. Ferris
- Department
of Physiology
and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Cody A. Siciliano
- Department
of Physiology
and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Sara R. Jones
- Department
of Physiology
and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| |
Collapse
|
10
|
Wu H, O'Neill B, Han DD, Thirtamara-Rajamani K, Wang Y, Gu HH. Restoration of cocaine stimulation and reward by reintroducing wild type dopamine transporter in adult knock-in mice with a cocaine-insensitive dopamine transporter. Neuropharmacology 2014; 86:31-7. [PMID: 24835281 DOI: 10.1016/j.neuropharm.2014.04.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 04/12/2014] [Accepted: 04/29/2014] [Indexed: 11/28/2022]
Abstract
In previous studies, we generated knock-in mice with a cocaine-insensitive dopamine transporter (DAT-CI mice) and found cocaine does not stimulate locomotion or produce reward in these mice, indicating DAT inhibition is necessary for cocaine stimulation and reward. However, DAT uptake is reduced in DAT-CI mice and thus the lack of cocaine responses could be due to adaptive changes. To test this, we used adeno-associated virus (AAV) to reintroduce the cocaine-sensitive wild type DAT (AAV-DATwt) back into adult DAT-CI mice, which restores cocaine inhibition of DAT in affected brain regions but does not reverse the adaptive changes. In an earlier study we showed that AAV-DATwt injections in regions covering the lateral nucleus accumbens (NAc) and lateral caudate-putamen (CPu) restored cocaine stimulation but not cocaine reward. In the current study, we expanded the AAV-DATwt infected areas to cover the olfactory tubercle (Tu) and the ventral midbrain (vMB) containing the ventral tegmental area (VTA) and substantia nigra (SN) in addition to CPu and NAc with multiple injections. These mice displayed the restoration of both locomotor stimulation and cocaine reward. We further found that AAV-DATwt injection in the vMB alone was sufficient to restore both cocaine stimulation and reward in DAT-CI mice. AAV injected in the VTA and SN resulted in DATwt expression and distribution to the DA terminal regions. In summary, cocaine induced locomotion and reward can be restored in fully developed DAT-CI mice, and cocaine inhibition of DAT expressed in dopaminergic neurons originated from the ventral midbrain mediates cocaine reward and stimulation.
Collapse
Affiliation(s)
- Haiyin Wu
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, Hubei, China; Department of Pharmacology, The Ohio State University, Columbus, OH 43210, USA
| | - Brian O'Neill
- Department of Pharmacology, The Ohio State University, Columbus, OH 43210, USA
| | - Dawn D Han
- Department of Pharmacology, The Ohio State University, Columbus, OH 43210, USA
| | | | - Yanlin Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, Hubei, China.
| | - Howard H Gu
- Department of Pharmacology, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
11
|
Calipari ES, Ferris MJ, Melchior JR, Bermejo K, Salahpour A, Roberts DCS, Jones SR. Methylphenidate and cocaine self-administration produce distinct dopamine terminal alterations. Addict Biol 2014; 19:145-55. [PMID: 22458761 DOI: 10.1111/j.1369-1600.2012.00456.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Methylphenidate (MPH) is a commonly abused psychostimulant prescribed for the treatment of attention deficit hyperactivity disorder. MPH has a mechanism of action similar to cocaine (COC) and is commonly characterized as a dopamine transporter (DAT) blocker. While there has been extensive work aimed at understanding dopamine (DA) nerve terminal changes following COC self-administration, very little is known about the effects of MPH self-administration on the DA system. We used fast scan cyclic voltammetry in nucleus accumbens core slices from animals with a 5-day self-administration history of 40 injections/day of either MPH (0.56 mg/kg) or COC (1.5 mg/kg) to explore alterations in baseline DA release and uptake kinetics as well as alterations in the interaction of each compound with the DAT. Although MPH and COC have similar behavioral effects, the consequences of self-administration on DA system parameters were found to be divergent. We show that COC self-administration reduced DAT levels and maximal rates of DA uptake, as well as reducing electrically stimulated release, suggesting decreased DA terminal function. In contrast, MPH self-administration increased DAT levels, DA uptake rates and DA release, suggesting enhanced terminal function, which was supported by findings of increased metabolite/DA tissue content ratios. Tyrosine hydroxylase messenger RNA, protein and phosphorylation levels were also assessed in both groups. Additionally, COC self-administration reduced COC-induced DAT inhibition, while MPH self-administration increased MPH-induced DAT inhibition, suggesting opposite pharmacodynamic effects of these two drugs. These findings suggest that the factors governing DA system adaptations are more complicated than simple DA uptake blockade.
Collapse
Affiliation(s)
- Erin S Calipari
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, USA Department of Pharmacology, University of Toronto, Toronto, Canada
| | | | | | | | | | | | | |
Collapse
|
12
|
Pramod AB, Foster J, Carvelli L, Henry LK. SLC6 transporters: structure, function, regulation, disease association and therapeutics. Mol Aspects Med 2013; 34:197-219. [PMID: 23506866 DOI: 10.1016/j.mam.2012.07.002] [Citation(s) in RCA: 209] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 07/03/2012] [Indexed: 02/08/2023]
Abstract
The SLC6 family of secondary active transporters are integral membrane solute carrier proteins characterized by the Na(+)-dependent translocation of small amino acid or amino acid-like substrates. SLC6 transporters, which include the serotonin, dopamine, norepinephrine, GABA, taurine, creatine, as well as amino acid transporters, are associated with a number of human diseases and disorders making this family a critical target for therapeutic development. In addition, several members of this family are directly involved in the action of drugs of abuse such as cocaine, amphetamines, and ecstasy. Recent advances providing structural insight into this family have vastly accelerated our ability to study these proteins and their involvement in complex biological processes.
Collapse
Affiliation(s)
- Akula Bala Pramod
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, United States
| | | | | | | |
Collapse
|
13
|
The membrane raft protein Flotillin-1 is essential in dopamine neurons for amphetamine-induced behavior in Drosophila. Mol Psychiatry 2013; 18:824-33. [PMID: 22710269 PMCID: PMC3582826 DOI: 10.1038/mp.2012.82] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The dopamine transporter (DAT) is the primary molecular target responsible for the rewarding properties of the psychostimulants amphetamine (AMPH) and cocaine. AMPH increases extracellular dopamine (DA) by promoting its nonexocytotic release via DAT-mediated efflux. Previous studies in heterologous cells have shown that phosphorylation of the amino terminus of DAT is required for AMPH-induced DA efflux but not for DA uptake. However, the identity of many of the modulatory proteins and the molecular mechanisms that coordinate efflux and the ensuing behavioral effects remain poorly defined. Here, we establish a robust assay for AMPH-induced hyperlocomotion in Drosophila melanogaster larvae. Using a variety of genetic and pharmacological approaches, we demonstrate that this behavioral response is dependent on DA and on DAT and its phosphorylation. We also show that methylphenidate (MPH), which competitively inhibits DA uptake but does not induce DAT-mediated DA efflux, also leads to DAT-dependent hyperlocomotion, but this response is independent of DAT phosphorylation. Moreover, we demonstrate that the membrane raft protein Flotillin-1 is required for AMPH-induced, but not MPH-induced, hyperlocomotion. These results are the first evidence of a role for a raft protein in an AMPH-mediated behavior. Thus, using our assay we are able to translate molecular and cellular findings to a behavioral level and to differentiate in vivo the distinct mechanisms of two psychostimulants.
Collapse
|
14
|
Schouw MLJ, De Ruiter MB, Kaag AM, van den Brink W, Lindauer RJL, Reneman L. Dopaminergic dysfunction in abstinent dexamphetamine users: results from a pharmacological fMRI study using a reward anticipation task and a methylphenidate challenge. Drug Alcohol Depend 2013; 130:52-60. [PMID: 23142493 DOI: 10.1016/j.drugalcdep.2012.10.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 10/10/2012] [Accepted: 10/15/2012] [Indexed: 11/15/2022]
Abstract
BACKGROUND Dopamine (DA) is involved in systems governing motor actions, motivational processes and cognitive functions. Preclinical studies have shown that even relatively low doses of d-amphetamine (dAMPH) (equivalent to doses used in clinical Practice) can lead to DA neurotoxicity in rodents and non-human primates (Ricaurte et al., 2005). METHODS Therefore, we investigated the DAergic function in eight male recreational users of dAMPH and eight male healthy controls using functional magnetic resonance imaging (fMRI). We compared brain activation between both groups during a monetary incentive delay task (Knutson et al., 2001) with and without an oral methylphenidate (MPH) challenge. All subjects were abstinent for at least 2 weeks during the baseline scan. The second scan was performed on the same day 1.5 h after receiving an oral dose of 35 mg MPH (approximately 0.5 mg/kg) when peak MPH binding was assumed. RESULTS When anticipating reward, dAMPH users showed lower striatal activation in comparison to control subjects. In addition, MPH induced a reduction in the striatal activation during reward anticipation in healthy controls, whereas no such effect was observed in dAMPH users. CONCLUSION The combination of these findings provides further evidence for frontostriatal DAergic dysfunction in recreational dAMPH users and is consistent with preclinical data suggesting neurotoxic effects of chronic dAMPH use. The findings of this explorative study could have important implications for humans in need for treatment with dAMPH, such as patients suffering from ADHD and therefore this study needs replication in a larger sample.
Collapse
Affiliation(s)
- M L J Schouw
- Department of Radiology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
15
|
Leo D, Gainetdinov RR. Transgenic mouse models for ADHD. Cell Tissue Res 2013; 354:259-71. [PMID: 23681253 PMCID: PMC3785710 DOI: 10.1007/s00441-013-1639-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 04/15/2013] [Indexed: 12/20/2022]
Abstract
Attention-deficit hyperactivity disorder (ADHD) is a developmental disorder characterized by symptoms of inattention, impulsivity and hyperactivity that adversely affect many aspects of life. Whereas the etiology of ADHD remains unknown, growing evidence indicates a genetic involvement in the development of this disorder. The brain circuits associated with ADHD are rich in monoamines, which are involved in the mechanism of action of psychostimulants and other medications used to treat this disorder. Dopamine (DA) is believed to play a major role in ADHD but other neurotransmitters are certainly also involved. Genetically modified mice have become an indispensable tool used to analyze the contribution of genetic factors in the pathogenesis of human disorders. Although rodent models cannot fully recapitulate complex human psychiatric disorders such as ADHD, transgenic mice offer an opportunity to directly investigate in vivo the specific roles of novel candidate genes identified in ADHD patients. Several knock-out and transgenic mouse models have been proposed as ADHD models, mostly based on targeting genes involved in DA transmission, including the gene encoding the dopamine transporter (DAT1). These mutant models provided an opportunity to evaluate the contribution of dopamine-related processes to brain pathology, to dissect the neuronal circuitry and molecular mechanisms involved in the antihyperkinetic action of psychostimulants and to evaluate novel treatments for ADHD. New transgenic models mouse models targeting other genes have recently been proposed for ADHD. Here, we discuss the recent advances and pitfalls in modeling ADHD endophenotypes in genetically altered animals.
Collapse
Affiliation(s)
- Damiana Leo
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Via Morego 30, Genoa, Italy,
| | | |
Collapse
|
16
|
O'Neill B, Tilley MR, Gu HH. Cocaine produces conditioned place aversion in mice with a cocaine-insensitive dopamine transporter. GENES BRAIN AND BEHAVIOR 2012; 12:34-8. [PMID: 23083326 DOI: 10.1111/j.1601-183x.2012.00872.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 09/13/2012] [Accepted: 10/17/2012] [Indexed: 01/27/2023]
Abstract
Cocaine is an inhibitor of the dopamine, norepinephrine and serotonin reuptake transporters. Because its administration would elevate signaling of all these three neurotransmitters, many studies have been aimed at attributing individual effects of cocaine to specific transmitter systems. Using mice with a cocaine-insensitive dopamine transporter (DAT-CI mice), we previously showed that cocaine-induced dopamine elevations were necessary for its rewarding and stimulating effects. In this study, we observe that DAT-CI mice exhibit cocaine-conditioned place aversion (CPA), and that its expression depends on their genetic background. Specifically, DAT-CI mice backcrossed to the C57Bl/6J strain background did not display a preference or an aversion to cocaine, whereas DAT-CI mice that were on a mixed 129S1/SvImJ × C57Bl/6J (129B6) background had a robust CPA to cocaine. These results indicate that while inhibition of the DAT is necessary for cocaine reward, other cocaine targets and neurotransmitter systems may mediate the aversive properties of cocaine. Furthermore, the aversive effect of cocaine can be observed in the absence of a DAT-mediated rewarding effect, and it is affected by genomic differences between these two mouse strains.
Collapse
Affiliation(s)
- B O'Neill
- Department of Pharmacology; Neuroscience Graduate Studies Program, The Ohio State University, Columbus, OH 43210, USA
| | | | | |
Collapse
|
17
|
Methylphenidate-elicited dopamine increases in ventral striatum are associated with long-term symptom improvement in adults with attention deficit hyperactivity disorder. J Neurosci 2012; 32:841-9. [PMID: 22262882 DOI: 10.1523/jneurosci.4461-11.2012] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Stimulant medications, such as methylphenidate, which are effective treatments for attention deficit hyperactivity disorder (ADHD), enhance brain dopamine signaling. However, the relationship between regional brain dopamine enhancement and treatment response has not been evaluated. Here, we assessed whether the dopamine increases elicited by methylphenidate are associated with long-term clinical response. We used a prospective design to study 20 treatment-naive adults with ADHD who were evaluated before treatment initiation and after 12 months of clinical treatment with a titrated regimen of oral methylphenidate. Methylphenidate-induced dopamine changes were evaluated with positron emission tomography and [(11)C]raclopride (D(2)/D(3) receptor radioligand sensitive to competition with endogenous dopamine). Clinical responses were assessed using the Conners' Adult ADHD Rating Scale and revealed a significant reduction in symptoms of inattention and hyperactivity with long-term methylphenidate treatment. A challenge dose of 0.5 mg/kg intravenous methylphenidate significantly increased dopamine in striatum (assessed as decreases in D(2)/D(3) receptor availability). In the ventral striatum, these dopamine increases were associated with the reductions in ratings of symptoms of inattention with clinical treatment. Statistical parametric mapping additionally showed dopamine increases in prefrontal and temporal cortices with intravenous methylphenidate that were also associated with decreases in symptoms of inattention. Our findings indicate that dopamine enhancement in ventral striatum (the brain region involved with reward and motivation) was associated with therapeutic response to methylphenidate, further corroborating the relevance of the dopamine reward/motivation circuitry in ADHD. It also provides preliminary evidence that methylphenidate-elicited dopamine increases in prefrontal and temporal cortices may also contribute to the clinical response.
Collapse
|
18
|
Castelli M, Federici M, Rossi S, De Chiara V, Napolitano F, Studer V, Motta C, Sacchetti L, Romano R, Musella A, Bernardi G, Siracusano A, Gu HH, Mercuri NB, Usiello A, Centonze D. Loss of striatal cannabinoid CB1 receptor function in attention-deficit / hyperactivity disorder mice with point-mutation of the dopamine transporter. Eur J Neurosci 2012; 34:1369-77. [PMID: 22034972 DOI: 10.1111/j.1460-9568.2011.07876.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Abnormal dopamine (DA) transmission in the striatum plays a pivotal role in attention-deficit/hyperactivity disorder (ADHD). As striatal DA signalling modulates the endocannabinoid system (ECS), the present study was aimed at investigating cannabinoid CB1 receptor (CB1R) function in a model of ADHD obtained by triple point-mutation in the dopamine transporter (DAT) gene in mice, making them insensitive to cocaine [DAT cocaine-insensitive (DAT-CI) mice]. DAT-CI mice had a marked hyperactive phenotype, and neurophysiological recordings revealed that the sensitivity of CB1Rs controlling GABA-mediated synaptic currents [CB1Rs((GABA)) ] in the striatum was completely lost. In contrast, CB1Rs modulating glutamate transmission [CB1Rs((Glu)) ], and GABA(B) receptors were not affected in this model of ADHD. In DAT-CI mice, the blockade of CB1R((GABA)) function was complete even after cocaine or environmental manipulations activating the endogenous DA-dependent reward system, which are known to sensitize these receptors in control animals. Conversely, the hedonic property of sucrose was intact in DAT-CI mice, indicating normal sweet perception in these animals. Our results point to CB1Rs as novel molecular players in ADHD, and suggest that therapeutic strategies aimed at interfering with the ECS might prove effective in this disorder.
Collapse
Affiliation(s)
- Maura Castelli
- Clinica Neurologica, Dipartimento di Neuroscienze, Università Tor Vergata, Via Montpellier, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Bracci E. The endocannabinoid system misfires in ADHD mice (commentary on Castelli et al.). Eur J Neurosci 2011; 34:1368. [PMID: 22034971 DOI: 10.1111/j.1460-9568.2011.07917.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Enrico Bracci
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
20
|
Archer T, Kostrzewa RM. Physical Exercise Alleviates ADHD Symptoms: Regional Deficits and Development Trajectory. Neurotox Res 2011; 21:195-209. [PMID: 21850535 DOI: 10.1007/s12640-011-9260-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 07/10/2011] [Accepted: 07/11/2011] [Indexed: 01/12/2023]
|
21
|
Negrao BL, Viljoen M. Stimulants and growth in children with attention-deficit/hyperactivity disorder. Med Hypotheses 2011; 77:21-8. [DOI: 10.1016/j.mehy.2011.03.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 02/22/2011] [Accepted: 03/07/2011] [Indexed: 10/18/2022]
|
22
|
Role of aberrant striatal dopamine D1 receptor/cAMP/protein kinase A/DARPP32 signaling in the paradoxical calming effect of amphetamine. J Neurosci 2010; 30:11043-56. [PMID: 20720111 DOI: 10.1523/jneurosci.1682-10.2010] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Attention deficit/hyperactivity disorder (ADHD) is characterized by inattention, impulsivity, and motor hyperactivity. Several lines of research support a crucial role for the dopamine transporter (DAT) gene in this psychiatric disease. Consistently, the most commonly prescribed medications in ADHD treatment are stimulant drugs, known to preferentially act on DAT. Recently, a knock-in mouse [DAT-cocaine insensitive (DAT-CI)] has been generated carrying a cocaine-insensitive DAT that is functional but with reduced dopamine uptake function. DAT-CI mutants display enhanced striatal extracellular dopamine levels and basal motor hyperactivity. Herein, we showed that DAT-CI animals present higher striatal dopamine turnover, altered basal phosphorylation state of dopamine and cAMP-regulated phosphoprotein 32 kDa (DARPP32) at Thr75 residue, but preserved D(2) receptor (D(2)R) function. However, although we demonstrated that striatal D(1) receptor (D(1)R) is physiologically responsive under basal conditions, its stimulus-induced activation strikingly resulted in paradoxical electrophysiological, behavioral, and biochemical responses. Indeed, in DAT-CI animals, (1) striatal LTP was completely disrupted, (2) R-(+)-6-chloro-7,8-dihydroxy-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepine hydrobromide (SKF 81297) treatment induced paradoxical motor calming effects, and (3) SKF 81297 administration failed to increase cAMP/protein kinase A (PKA)/DARPP32 signaling. Such biochemical alteration selectively affected dopamine D(1)Rs since haloperidol, by blocking the tonic inhibition of D(2)R, unmasked a normal activation of striatal adenosine A(2A) receptor-mediated cAMP/PKA/DARPP32 cascade in mutants. Most importantly, our studies highlighted that amphetamine, nomifensine, and bupropion, through increased striatal dopaminergic transmission, are able to revert motor hyperactivity of DAT-CI animals. Overall, our results suggest that the paradoxical motor calming effect induced by these drugs in DAT-CI mutants depends on selective aberrant phasic activation of D(1)R/cAMP/PKA/DARPP32 signaling in response to increased striatal extracellular dopamine levels.
Collapse
|
23
|
Krapacher FA, Mlewski EC, Ferreras S, Pisano V, Paolorossi M, Hansen C, Paglini G. Mice lacking p35 display hyperactivity and paradoxical response to psychostimulants. J Neurochem 2010; 114:203-14. [PMID: 20403084 DOI: 10.1111/j.1471-4159.2010.06748.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cyclin-dependent kinase 5/p35 kinase complex plays a critical role in dopaminergic neurotransmission. Dysregulation of dopamine (DA) signaling is associated with neurological and neuropsychiatric disorders. As cyclin-dependent kinase 5 (Cdk5) requires association with p35 for its proper activation, we hypothesized that dysregulation of Cdk5 activity might have an effect on striatal-mediated behavior. We used a mutant mouse, deficient in p35 protein (p35 KO), which displayed reduced Cdk5 activity. Throughout behavioral and biochemical characterization of naïve and psychostimulant-treated mice, we demonstrated that only juvenile p35 KO mice displayed spontaneous hyperactivity, responded with a paradoxical hypolocomotor effect to psychostimulant drugs and exhibited deficit on proper behavioral inhibition. Strong immunolabeling for tyrosine-hydroxylase and high striatal DA synthesis and contents with a low DA turnover, which were reverted by psychostimulants, were also found in mutant mice. Our results demonstrate that p35 deficiency is critically involved in the expression of a hyperactive behavioral phenotype with hyper-functioning of the dopaminergic system, emphasizing the importance of proper Cdk5 kinase activity for normal motor and emotional features. Thus, p35 KO mice may be another useful animal model for understanding cellular and molecular events underlying attention deficit hyperactivity disorder-like disorders.
Collapse
Affiliation(s)
- Favio Ariel Krapacher
- Laboratory of Neurobiology and Cell Biology, Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC-CONICET), 5016 Córdoba, Argentina
| | | | | | | | | | | | | |
Collapse
|
24
|
Sora I, Li B, Igari M, Hall FS, Ikeda K. Transgenic mice in the study of drug addiction and the effects of psychostimulant drugs. Ann N Y Acad Sci 2010; 1187:218-46. [PMID: 20201856 DOI: 10.1111/j.1749-6632.2009.05276.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The first transgenic models used to study addiction were based upon a priori assumptions about the importance of particular genes in addiction, including the main target molecules of morphine, amphetamine, and cocaine. This consequently emphasized the importance of monoamine transporters, opioid receptors, and monoamine receptors in addiction. Although the effects of opiates were largely eliminated by mu opioid receptor gene knockout, the case for psychostimulants was much more complex. Research using transgenic models supported the idea of a polygenic basis for psychostimulant effects and has associated particular genes with different behavioral consequences of psychostimulants. Phenotypic analysis of transgenic mice, especially gene knockout mice, has been instrumental in identifying the role of specific molecular targets of addictive drugs in their actions. In this article, we summarize studies that have provided insight into the polygenic determination of drug addiction phenotypes in ways that are not possible with other methods, emphasizing research into the effects of psychostimulant drugs in gene knockouts of the monoamine transporters and monoamine receptors.
Collapse
Affiliation(s)
- Ichiro Sora
- Department of Biological Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | | | | | | | | |
Collapse
|
25
|
Monoaminergic neuronal changes in orexin deficient mice. Neuropharmacology 2010; 58:826-32. [DOI: 10.1016/j.neuropharm.2009.08.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2009] [Revised: 08/15/2009] [Accepted: 08/17/2009] [Indexed: 11/17/2022]
|
26
|
Issy A, Salum C, Del Bel E. Nitric oxide modulation of methylphenidate-induced disruption of prepulse inhibition in Swiss mice. Behav Brain Res 2009; 205:475-81. [DOI: 10.1016/j.bbr.2009.08.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Revised: 07/29/2009] [Accepted: 08/03/2009] [Indexed: 11/24/2022]
|