1
|
Savchenko A, Targa G, Fesenko Z, Leo D, Gainetdinov RR, Sukhanov I. Dopamine Transporter Deficient Rodents: Perspectives and Limitations for Neuroscience. Biomolecules 2023; 13:806. [PMID: 37238676 PMCID: PMC10216310 DOI: 10.3390/biom13050806] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
The key element of dopamine (DA) neurotransmission is undoubtedly DA transporter (DAT), a transmembrane protein responsible for the synaptic reuptake of the mediator. Changes in DAT's function can be a key mechanism of pathological conditions associated with hyperdopaminergia. The first strain of gene-modified rodents with a lack of DAT were created more than 25 years ago. Such animals are characterized by increased levels of striatal DA, resulting in locomotor hyperactivity, increased levels of motor stereotypes, cognitive deficits, and other behavioral abnormalities. The administration of dopaminergic and pharmacological agents affecting other neurotransmitter systems can mitigate those abnormalities. The main purpose of this review is to systematize and analyze (1) known data on the consequences of changes in DAT expression in experimental animals, (2) results of pharmacological studies in these animals, and (3) to estimate the validity of animals lacking DAT as models for discovering new treatments of DA-related disorders.
Collapse
Affiliation(s)
- Artem Savchenko
- Valdman Institute of Pharmacology, Pavlov First St. Petersburg State Medical University, Lev Tolstoy Str. 6-8, 197022 St. Petersburg, Russia;
| | - Giorgia Targa
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy
| | - Zoia Fesenko
- Institute of Translational Biomedicine, St. Petersburg State University, 7/9 Universitetskaya Emb., 199034 St. Petersburg, Russia
| | - Damiana Leo
- Department of Neurosciences, University of Mons, 7000 Mons, Belgium
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, 7/9 Universitetskaya Emb., 199034 St. Petersburg, Russia
- St. Petersburg University Hospital, St. Petersburg State University, Fontanka River Emb. 154, 190121 St. Petersburg, Russia
| | - Ilya Sukhanov
- Valdman Institute of Pharmacology, Pavlov First St. Petersburg State Medical University, Lev Tolstoy Str. 6-8, 197022 St. Petersburg, Russia;
- St. Petersburg University Hospital, St. Petersburg State University, Fontanka River Emb. 154, 190121 St. Petersburg, Russia
| |
Collapse
|
2
|
Sotoyama H, Namba H, Tohmi M, Nawa H. Schizophrenia Animal Modeling with Epidermal Growth Factor and Its Homologs: Their Connections to the Inflammatory Pathway and the Dopamine System. Biomolecules 2023; 13:biom13020372. [PMID: 36830741 PMCID: PMC9953688 DOI: 10.3390/biom13020372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/10/2023] [Accepted: 02/12/2023] [Indexed: 02/18/2023] Open
Abstract
Epidermal growth factor (EGF) and its homologs, such as neuregulins, bind to ErbB (Her) receptor kinases and regulate glial differentiation and dopaminergic/GABAergic maturation in the brain and are therefore implicated in schizophrenia neuropathology involving these cell abnormalities. In this review, we summarize the biological activities of the EGF family and its neuropathologic association with schizophrenia, mainly overviewing our previous model studies and the related articles. Transgenic mice as well as the rat/monkey models established by perinatal challenges of EGF or its homologs consistently exhibit various behavioral endophenotypes relevant to schizophrenia. In particular, post-pubertal elevation in baseline dopaminergic activity may illustrate the abnormal behaviors relevant to positive and negative symptoms as well as to the timing of this behavioral onset. With the given molecular interaction and transactivation of ErbB receptor kinases with Toll-like receptors (TLRs), EGF/ErbB signals are recruited by viral infection and inflammatory diseases such as COVID-19-mediated pneumonia and poxvirus-mediated fibroma and implicated in the immune-inflammatory hypothesis of schizophrenia. Finally, we also discuss the interaction of clozapine with ErbB receptor kinases as well as new antipsychotic development targeting these receptors.
Collapse
Affiliation(s)
- Hidekazu Sotoyama
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
- Department of Physiology, School of Medicine, Niigata University, Niigata 951-8122, Japan
- Correspondence: (H.N.); (H.S.)
| | - Hisaaki Namba
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama 649-8156, Japan
| | - Manavu Tohmi
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama 649-8156, Japan
| | - Hiroyuki Nawa
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama 649-8156, Japan
- Correspondence: (H.N.); (H.S.)
| |
Collapse
|
3
|
Cid-Jofré V, Moreno M, Sotomayor-Zárate R, Cruz G, Renard GM. Modafinil Administration to Preadolescent Rat Impairs Non-Selective Attention, Frontal Cortex D 2 Expression and Mesolimbic GABA Levels. Int J Mol Sci 2022; 23:ijms23126602. [PMID: 35743046 PMCID: PMC9223864 DOI: 10.3390/ijms23126602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/26/2022] [Accepted: 06/09/2022] [Indexed: 11/16/2022] Open
Abstract
The misuse of psychostimulants is an increasing behavior among young people, highlighting in some countries the abuse of modafinil (MOD) as a neuropotentiator. However, several clinical trials are investigating MOD as an alternative pharmacological treatment for attentional deficit and hyperactivity disorder (ADHD) in children and adolescents. On the other hand, the early use of psychostimulants and the misdiagnosis rates in ADHD make it crucial to investigate the brain effects of this type of drug in young healthy individuals. The aim of this work was to evaluate the effects of chronic MOD treatment on neurochemicals (γ-aminobutyric acid and glutamate), dopamine receptor 2 (D2) expression and behavior (non-selective attention "NSA") in the mesocorticolimbic system of young healthy Sprague-Dawley rats. Preadolescent male rats were injected with MOD (75 mg/kg, i.p.) or a vehicle for 14 days (from postnatal day 22 to 35). At postnatal day 36, we measured the GLU and GABA contents and their extracellular levels in the nucleus accumbens (NAc). In addition, the GLU and GABA contents were measured in the ventral tegmental area (VTA) and D2 protein levels in the prefrontal cortex (PFC). Chronic use of MOD during adolescence induces behavioral and neurochemical changes associated with the mesocorticolimbic system, such as a reduction in PFC D2 expression, VTA GABA levels and NSA. These results contribute to the understanding of the neurological effects of chronic MOD use on a young healthy brain.
Collapse
Affiliation(s)
- Valeska Cid-Jofré
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Obispo Umaña 050, Estación Central, Santiago 9160019, Chile; (V.C.-J.); (M.M.)
| | - Macarena Moreno
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Obispo Umaña 050, Estación Central, Santiago 9160019, Chile; (V.C.-J.); (M.M.)
- Escuela de Psicología, Facultad de Ciencias Sociales, Universidad Bernardo O’Higgins, Santiago 8370993, Chile
| | - Ramón Sotomayor-Zárate
- Laboratorio de Neuroquímica y Neurofarmacología, Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Av. Gran Bretaña 1111, Playa Ancha, Valparaíso 2360102, Chile;
| | - Gonzalo Cruz
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Av. Gran Bretaña 1111, Playa Ancha, Valparaíso 2360102, Chile;
| | - Georgina M. Renard
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Obispo Umaña 050, Estación Central, Santiago 9160019, Chile; (V.C.-J.); (M.M.)
- Correspondence:
| |
Collapse
|
4
|
Sotoyama H, Inaba H, Iwakura Y, Namba H, Takei N, Sasaoka T, Nawa H. The dual role of dopamine in the modulation of information processing in the prefrontal cortex underlying social behavior. FASEB J 2022; 36:e22160. [DOI: 10.1096/fj.202101637r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/12/2021] [Accepted: 12/29/2021] [Indexed: 01/08/2023]
Affiliation(s)
- Hidekazu Sotoyama
- Department of Molecular Neurobiology Brain Research Institute, Niigata University Niigata Japan
| | - Hiroyoshi Inaba
- Department of Molecular Neurobiology Brain Research Institute, Niigata University Niigata Japan
| | - Yuriko Iwakura
- Department of Molecular Neurobiology Brain Research Institute, Niigata University Niigata Japan
- Department of Brain Tumor Biology Brain Research Institute, Niigata University Niigata Japan
| | - Hisaaki Namba
- Department of Molecular Neurobiology Brain Research Institute, Niigata University Niigata Japan
- Department of Physiological Sciences, School of Pharmaceutical Sciences Wakayama Medical University Wakayama Japan
| | - Nobuyuki Takei
- Department of Molecular Neurobiology Brain Research Institute, Niigata University Niigata Japan
- Department of Brain Tumor Biology Brain Research Institute, Niigata University Niigata Japan
| | - Toshikuni Sasaoka
- Department of Comparative & Experimental Medicine Brain Research Institute, Niigata University Niigata Japan
| | - Hiroyuki Nawa
- Department of Molecular Neurobiology Brain Research Institute, Niigata University Niigata Japan
- Department of Physiological Sciences, School of Pharmaceutical Sciences Wakayama Medical University Wakayama Japan
| |
Collapse
|
5
|
Beeler JA, Burghardt NS. The Rise and Fall of Dopamine: A Two-Stage Model of the Development and Entrenchment of Anorexia Nervosa. Front Psychiatry 2022; 12:799548. [PMID: 35087433 PMCID: PMC8787068 DOI: 10.3389/fpsyt.2021.799548] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/14/2021] [Indexed: 12/03/2022] Open
Abstract
Dopamine has long been implicated as a critical neural substrate mediating anorexia nervosa (AN). Despite nearly 50 years of research, the putative direction of change in dopamine function remains unclear and no consensus on the mechanistic role of dopamine in AN has been achieved. We hypothesize two stages in AN- corresponding to initial development and entrenchment- characterized by opposite changes in dopamine. First, caloric restriction, particularly when combined with exercise, triggers an escalating spiral of increasing dopamine that facilitates the behavioral plasticity necessary to establish and reinforce weight-loss behaviors. Second, chronic self-starvation reverses this escalation to reduce or impair dopamine which, in turn, confers behavioral inflexibility and entrenchment of now established AN behaviors. This pattern of enhanced, followed by impaired dopamine might be a common path to many behavioral disorders characterized by reinforcement learning and subsequent behavioral inflexibility. If correct, our hypothesis has significant clinical and research implications for AN and other disorders, such as addiction and obesity.
Collapse
Affiliation(s)
- Jeff A. Beeler
- Department of Psychology, Queens College, City University of New York, Flushing, NY, United States
- Psychology Program, The Graduate Center, CUNY, New York, NY, United States
- Biology Program, The Graduate Center, City University of New York, New York, NY, United States
| | - Nesha S. Burghardt
- Psychology Program, The Graduate Center, CUNY, New York, NY, United States
- Department of Psychology, Hunter College, CUNY, New York, NY, United States
| |
Collapse
|
6
|
Savchenko A, Müller C, Lubec J, Leo D, Korz V, Afjehi-Sadat L, Malikovic J, Sialana FJ, Lubec G, Sukhanov I. The Lack of Dopamine Transporter Is Associated With Conditional Associative Learning Impairments and Striatal Proteomic Changes. Front Psychiatry 2022; 13:799433. [PMID: 35370807 PMCID: PMC8971526 DOI: 10.3389/fpsyt.2022.799433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/04/2022] [Indexed: 11/14/2022] Open
Abstract
Dopamine (DA) is critically involved in different functions of the central nervous system (CNS) including control of voluntary movement, affect, reward, sleep, and cognition. One of the key components of DA neurotransmission is DA reuptake by the DA transporter (DAT), ensuring rapid clearance of DA from the synaptic cleft. Thus, lack of DAT leads to persistent high extracellular DA levels. While there is strong evidence for a role of striatal dopaminergic activity in learning and memory processes, little is known about the contribution of DAT deficiency to conditional learning impairments and underlying molecular processes. DAT-knockout (DAT-KO) rats were tested in a set of behavioral experiments evaluating conditional associative learning, which requires unaltered striatal function. In parallel, a large-scale proteomic analysis of the striatum was performed to identify molecular factors probably underlying behavioral patterns. DAT-KO rats were incapable to acquire a new operant skill in Pavlovian/instrumental autoshaping, although the conditional stimulus-unconditional stimulus (CS-US) association seems to be unaffected. These findings suggest that DAT directly or indirectly contributes to the reduction of transference of incentive salience from the reward to the CS. We propose that specific impairment of conditional learning might be caused by molecular adaptations to the hyperdopaminergic state, presumably by dopamine receptor 1 (DRD1) hypofunction, as proposed by proteomic analysis. Whether DRD1 downregulation can cause cognitive deficits in the hyperdopaminergic state is the subject of discussion, and further studies are needed to answer this question. This study may be useful for the interpretation of previous and the design of future studies in the dopamine field.
Collapse
Affiliation(s)
- Artem Savchenko
- Institute of Pharmacology, Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia
| | - Carina Müller
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Jana Lubec
- Programme for Proteomics, Paracelsus Medical University, Salzburg, Austria
| | - Damiana Leo
- Department of Neurosciences, University of Mons, Mons, Belgium
| | - Volker Korz
- Programme for Proteomics, Paracelsus Medical University, Salzburg, Austria
| | - Leila Afjehi-Sadat
- Programme for Proteomics, Paracelsus Medical University, Salzburg, Austria
| | - Jovana Malikovic
- Programme for Proteomics, Paracelsus Medical University, Salzburg, Austria
| | - Fernando J Sialana
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Gert Lubec
- Programme for Proteomics, Paracelsus Medical University, Salzburg, Austria
| | - Ilya Sukhanov
- Institute of Pharmacology, Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia
| |
Collapse
|
7
|
Timing behavior in genetic murine models of neurological and psychiatric diseases. Exp Brain Res 2021; 239:699-717. [PMID: 33404792 DOI: 10.1007/s00221-020-06021-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 12/16/2020] [Indexed: 01/17/2023]
Abstract
How timing behavior is altered in different neurodevelopmental and neurodegenerative disorders is a contemporary research question. Genetic murine models (GMM) that offer high construct validity also serve as useful tools to investigate this question. But the literature on timing behavior of different GMMs largely remains to be consolidated. The current paper addresses this gap by reviewing studies that have been conducted with GMMs of neurodevelopmental (e.g. ADHD, schizophrenia, autism spectrum disorder), neurodegenerative disorders (e.g., Alzheimer's disease, Huntington's disease) as well as circadian and other mutant lines. The review focuses on those studies that specifically utilized the peak interval procedure to improve the comparability of findings both within and between different disease models. The reviewed studies revealed timing deficits that are characteristic of different disorders. Specifically, Huntington's disease models had weaker temporal control over the termination of their anticipatory responses, Alzheimer's disease models had earlier timed responses, schizophrenia models had weaker temporal control, circadian mutants had shifted timed responses consistent with shifts in the circadian periods. The differences in timing behavior were less consistent for other conditions such as attention deficit and hyperactivity disorder and mutations related to intellectual disability. We discuss the implications of these findings for the neural basis of an internal stopwatch. Finally, we make methodological recommendations for future research for improving the comparability of the timing behavior across different murine models.
Collapse
|
8
|
Enhanced Dopamine Transmission and Hyperactivity in the Dopamine Transporter Heterozygous Mice Lacking the D3 Dopamine Receptor. Int J Mol Sci 2020; 21:ijms21218216. [PMID: 33153031 PMCID: PMC7662256 DOI: 10.3390/ijms21218216] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 12/29/2022] Open
Abstract
Dopamine transporter knockout (DATk) mice are known to demonstrate profound hyperactivity concurrent with elevated (5-fold) extracellular dopamine in the basal ganglia. At the same time, heterozygous DAT mice (DATh) demonstrate a 2-fold increase in dopamine levels yet only a marginal elevation in locomotor activity level. Another model of dopaminergic hyperactivity is the D3 dopamine receptor knockout (D3k) mice, which present only a modest hyperactivity phenotype, predominately manifested as stereotypical behaviors. In the D3k mice, the hyperactivity is also correlated with elevated extracellular dopamine levels (2-fold) in the basal ganglia. Cross-breeding was used to evaluate the functional consequences of the deletion of both genes. In the heterozygous DAT mice, inactivation of the D3R gene (DATh/D3k) resulted in significant hyperactivity and further elevation of striatal extracellular dopamine above levels observed in respective single mutant mice. The decreased weight of DATk mice was evident regardless of the D3 dopamine receptor genotype. In contrast, measures of thermoregulation revealed that the marked hypothermia of DATk mice (−2 °C) was reversed in double knockout mice. Thus, the extracellular dopamine levels elevated by prolonging uptake could be elevated even further by eliminating the D3 receptor. These data also suggest that the hypothermia observed in DATk mice may be mediated through D3 receptors.
Collapse
|
9
|
Sousa A, Dinis-Oliveira RJ. Pharmacokinetic and pharmacodynamic of the cognitive enhancer modafinil: Relevant clinical and forensic aspects. Subst Abus 2020; 41:155-173. [PMID: 31951804 DOI: 10.1080/08897077.2019.1700584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Modafinil is a nonamphetamine nootropic drug with an increasingly therapeutic interest due to its different sites of action and behavioral effects in comparison to cocaine or amphetamine. A review of modafinil (and of its prodrug adrafinil and its R-enantiomer armodafinil) chemical, pharmacokinetic, pharmacodynamic, toxicological, clinical and forensic aspects was performed, aiming to better understand possible health problems associated to its unconscious and unruled use. Modafinil is a racemate metabolized mainly in the liver into its inactive acid and sulfone metabolites, which undergo primarily renal excretion. Although not fully clarified, major effects seem to be associated to inhibition of dopamine reuptake and modulation of several other neurochemical pathways, namely noradrenergic, serotoninergic, orexinergic, histaminergic, glutamatergic and GABAergic. Due its wake-promoting effects, modafinil is used for the treatment of daily sleepiness associated to narcolepsy, obstructive sleep apnea and shift work sleep disorder. Its psychotropic and cognitive effects are also attractive in several other pathologies and conditions that affect sleep structure, induce fatigue and lethargy, and impair cognitive abilities. Additionally, in health subjects, including students, modafinil is being used off-label to overcome sleepiness, increase concentration and improve cognitive potential. The most common adverse effects associated to modafinil intake are headache, insomnia, anxiety, diarrhea, dry mouth and raise in blood pressure and heart rate. Infrequently, severe dermatologic effects in children, including maculopapular and morbilliform rash, erythema multiforme and Stevens-Johnson Syndrome have been reported. Intoxication and dependence associated to modafinil are uncommon. Further research on effects and health implications of modafinil and its analogs is steel needed to create evidence-based policies.
Collapse
Affiliation(s)
- Ana Sousa
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Ricardo Jorge Dinis-Oliveira
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, Porto, Portugal.,IINFACTS - Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, Gandra, Portugal.,UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
10
|
Gene knockout animal models of depression, anxiety and obsessive compulsive disorders. Psychiatr Genet 2019; 29:191-199. [DOI: 10.1097/ypg.0000000000000238] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
11
|
Kim M, Custodio RJ, Botanas CJ, de la Peña JB, Sayson LV, Abiero A, Ryoo ZY, Cheong JH, Kim HJ. The circadian gene, Per2, influences methamphetamine sensitization and reward through the dopaminergic system in the striatum of mice. Addict Biol 2019; 24:946-957. [PMID: 30091820 DOI: 10.1111/adb.12663] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 06/17/2018] [Accepted: 06/27/2018] [Indexed: 01/19/2023]
Abstract
Drug addiction is a chronic and relapsing brain disorder, influenced by complex interactions between endogenous and exogenous factors. Per2, a circadian gene, plays a role in drug addiction. Previous studies using Per2-knockout mice have shown a role for Per2 in cocaine, morphine and alcohol addiction. In the present study, we investigated the role of Per2 in methamphetamine (METH) addiction using Per2-overexpression and knockout mice. We observed locomotor sensitization responses to METH administration, and rewarding effects using a conditioned place preference test. In addition, we measured expression levels of dopamine and dopamine-related genes (monoamine oxidase A, DA receptor 1, DA receptor 2, DA active transporter, tyrosine hydroxylase and cAMP response element-binding protein 1) in the striatum of the mice after repeated METH treatments, using qRT-PCR. Per2-overexpressed mice showed decreased locomotor sensitization and rewarding effects of METH compared to the wildtype mice, whereas the opposite was observed in Per2 knockout mice. Both types of transgenic mice showed altered expression levels of dopamine-related genes after repeated METH administration. Specifically, we observed lower dopamine levels in Per2-overexpressed mice and higher levels in Per2-knockout mice. Taken together, Per2 expression levels may influence the addictive effects of METH through the dopaminergic system in the striatum of mice.
Collapse
Affiliation(s)
- Mikyung Kim
- Uimyung Research Institute for Neuroscience, Department of Pharmacy; Sahmyook University; Korea
| | - Raly James Custodio
- Uimyung Research Institute for Neuroscience, Department of Pharmacy; Sahmyook University; Korea
| | - Chrislean Jun Botanas
- Uimyung Research Institute for Neuroscience, Department of Pharmacy; Sahmyook University; Korea
| | | | - Leandro Val Sayson
- Uimyung Research Institute for Neuroscience, Department of Pharmacy; Sahmyook University; Korea
| | - Arvie Abiero
- Uimyung Research Institute for Neuroscience, Department of Pharmacy; Sahmyook University; Korea
| | - Zae Young Ryoo
- School of Life Science, BK21 Plus KNU Creative Bio Research Group, College of Natural Sciences; Kyungpook National University; Korea
| | - Jae Hoon Cheong
- Uimyung Research Institute for Neuroscience, Department of Pharmacy; Sahmyook University; Korea
| | - Hee Jin Kim
- Uimyung Research Institute for Neuroscience, Department of Pharmacy; Sahmyook University; Korea
| |
Collapse
|
12
|
Li Q, Jia Y, Burris WR, Bridges PJ, Matthews JC. Forms of selenium in vitamin-mineral mixes differentially affect the expression of genes responsible for prolactin, ACTH, and α-MSH synthesis and mitochondrial dysfunction in pituitaries of steers grazing endophyte-infected tall fescue. J Anim Sci 2019; 97:631-643. [PMID: 30476104 DOI: 10.1093/jas/sky438] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 08/14/2018] [Indexed: 01/08/2023] Open
Abstract
The goal of this study was to test the hypothesis that sodium selenite (inorganic Se, ISe), SEL-PLEX (organic forms of Se, OSe), vs. a 1:1 blend (MIX) of ISe and OSe in a basal vitamin-mineral (VM) mix would differentially alter pituitary transcriptome profiles in growing beef steers grazing an endophyte-infected tall fescue (E+) pasture. Predominately Angus steers (BW = 183 ± 34 kg) were randomly selected from fall-calving cows grazing E+ pasture and consuming VM mixes that contained 35 ppm Se as ISe, OSe, or MIX forms. Steers were weaned, depleted of Se for 98 d, and subjected to summer-long common grazing of a 10.1 ha E+ pasture containing 0.51 ppm ergot alkaloids. Steers were assigned (n = 8 per treatment) to the same Se-form treatments on which they were raised. Selenium treatments were administered by daily top-dressing 85 g of VM mix onto 0.23 kg soyhulls, using in-pasture Calan gates. As previously reported, serum prolactin was greater for MIX (52%) and OSe (59%) steers vs. ISe. Pituitaries were collected at slaughter and changes in global and selected mRNA expression patterns determined by microarray and real-time reverse transcription PCR analyses, respectively. The effects of Se treatment on relative gene expression were subjected to one-way ANOVA. The form of Se affected the expression of 542 annotated genes (P < 0.005). Integrated pathway analysis found a canonical pathway network between prolactin and pro-opiomelanocortin (POMC)/ACTH/α-melanocyte-stimulating hormone (α-MSH) synthesis-related proteins and that mitochondrial dysfunction was a top-affected canonical pathway. Targeted reverse transcription-PCR analysis found that the relative abundance of mRNA encoding prolactin and POMC/ACTH/α-MSH synthesis-related proteins was affected (P < 0.05) by the form of Se, as were (P ≤ 0.05) mitochondrial dysfunction-related proteins (CYB5A, FURIN, GPX4, and PSENEN). OSe steers appeared to have a greater prolactin synthesis capacity (more PRL mRNA) vs. ISe steers through decreased dopamine type two receptor signaling (more DRD2 mRNA), whereas MIX steers had a greater prolactin synthesis capacity (more PRL mRNA) and release potential by increasing thyrotropin-releasing hormone concentrations (less TRH receptor mRNA) than ISe steers. OSe steers also had a greater ACTH and α-MSH synthesis potential (more POMC, PCSK2, CPE, and PAM mRNA) than ISe steers. We conclude that form of Se in VM mixes altered expression of genes responsible for prolactin and POMC/ACTH/α-MSH synthesis, and mitochondrial function, in pituitaries of growing beef steers subjected to summer-long grazing an E+ pasture.
Collapse
Affiliation(s)
- Qing Li
- Department of Animal and Food Sciences, University of Kentucky, Lexington, Kentucky
| | - Yang Jia
- Department of Animal and Food Sciences, University of Kentucky, Lexington, Kentucky
| | - Walter R Burris
- Department of Animal and Food Sciences, University of Kentucky, Lexington, Kentucky
| | - Phillip J Bridges
- Department of Animal and Food Sciences, University of Kentucky, Lexington, Kentucky
| | - James C Matthews
- Department of Animal and Food Sciences, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
13
|
Solinas M, Belujon P, Fernagut PO, Jaber M, Thiriet N. Dopamine and addiction: what have we learned from 40 years of research. J Neural Transm (Vienna) 2018; 126:481-516. [PMID: 30569209 DOI: 10.1007/s00702-018-1957-2] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 11/17/2018] [Indexed: 12/22/2022]
Abstract
Among the neurotransmitters involved in addiction, dopamine (DA) is clearly the best known. The critical role of DA in addiction is supported by converging evidence that has been accumulated in the last 40 years. In the present review, first we describe the dopaminergic system in terms of connectivity, functioning and involvement in reward processes. Second, we describe the functional, structural, and molecular changes induced by drugs within the DA system in terms of neuronal activity, synaptic plasticity and transcriptional and molecular adaptations. Third, we describe how genetic mouse models have helped characterizing the role of DA in addiction. Fourth, we describe the involvement of the DA system in the vulnerability to addiction and the interesting case of addiction DA replacement therapy in Parkinson's disease. Finally, we describe how the DA system has been targeted to treat patients suffering from addiction and the result obtained in clinical settings and we discuss how these different lines of evidence have been instrumental in shaping our understanding of the physiopathology of drug addiction.
Collapse
Affiliation(s)
- Marcello Solinas
- Université de Poitiers, INSERM, U-1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France.
| | - Pauline Belujon
- Université de Poitiers, INSERM, U-1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Pierre Olivier Fernagut
- Université de Poitiers, INSERM, U-1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| | - Mohamed Jaber
- Université de Poitiers, INSERM, U-1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
- CHU de Poitiers, Poitiers, France
| | - Nathalie Thiriet
- Université de Poitiers, INSERM, U-1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France
| |
Collapse
|
14
|
de Matos LO, Reis ALDAL, Guerra LTL, Guarnieri LDO, Moraes MA, Aquino NSS, Szawka RE, Pereira GS, Souza BR. l-Dopa treatment during perinatal development leads to different behavioral alterations in female vs. male juvenile Swiss mice. Pharmacol Biochem Behav 2018; 173:1-14. [DOI: 10.1016/j.pbb.2018.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/25/2018] [Accepted: 08/06/2018] [Indexed: 12/26/2022]
|
15
|
Efimova EV, Gainetdinov RR, Budygin EA, Sotnikova TD. Dopamine transporter mutant animals: a translational perspective. J Neurogenet 2017; 30:5-15. [PMID: 27276191 DOI: 10.3109/01677063.2016.1144751] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The dopamine transporter (DAT) plays an important homeostatic role in the control of both the extracellular and intraneuronal concentrations of dopamine, thereby providing effective control over activity of dopaminergic transmission. Since brain dopamine is known to be involved in numerous neuropsychiatric disorders, investigations using mice with genetically altered DAT function and thus intensity of dopamine-mediated signaling have provided numerous insights into the pathology of these disorders and novel pathological mechanisms that could be targeted to provide new therapeutic approaches for these disorders. In this brief overview, we discuss recent investigations involving animals with genetically altered DAT function, particularly focusing on translational studies providing new insights into pathology and pharmacology of dopamine-related disorders. Perspective applications of these and newly developed models of DAT dysfunction are also discussed.
Collapse
Affiliation(s)
- Evgeniya V Efimova
- a Institute of Translational Biomedicine, St. Petersburg State University , St. Petersburg , Russia ;,b Skolkovo Institute of Science and Technology , Skolkovo , Moscow Region , Russia
| | - Raul R Gainetdinov
- a Institute of Translational Biomedicine, St. Petersburg State University , St. Petersburg , Russia ;,b Skolkovo Institute of Science and Technology , Skolkovo , Moscow Region , Russia
| | - Evgeny A Budygin
- a Institute of Translational Biomedicine, St. Petersburg State University , St. Petersburg , Russia ;,c Department of Neurobiology and Anatomy , Wake Forest School of Medicine , Winston-Salem , NC , USA
| | - Tatyana D Sotnikova
- a Institute of Translational Biomedicine, St. Petersburg State University , St. Petersburg , Russia
| |
Collapse
|
16
|
Pittenger C. Histidine Decarboxylase Knockout Mice as a Model of the Pathophysiology of Tourette Syndrome and Related Conditions. Handb Exp Pharmacol 2017; 241:189-215. [PMID: 28233179 PMCID: PMC5538774 DOI: 10.1007/164_2016_127] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
While the normal functions of histamine (HA) in the central nervous system have gradually come into focus over the past 30 years, the relationship of abnormalities in neurotransmitter HA to human disease has been slower to emerge. New insight came with the 2010 description of a rare nonsense mutation in the biosynthetic enzyme histidine decarboxylase (Hdc) that was associated with Tourette syndrome (TS) and related conditions in a single family pedigree. Subsequent genetic work has provided further support for abnormalities of HA signaling in sporadic TS. As a result of this genetic work, Hdc knockout mice, which were generated more than 15 years ago, have been reexamined as a model of the pathophysiology of TS and related conditions. Parallel work in these KO mice and in human carriers of the Hdc mutation has revealed abnormalities in the basal ganglia system and its modulation by dopamine (DA) and has confirmed the etiologic, face, and predictive validity of the model. The Hdc-KO model thus serves as a unique platform to probe the pathophysiology of TS and related conditions, and to generate specific hypotheses for subsequent testing in humans. This chapter summarizes the development and validation of this model and recent and ongoing work using it to further investigate pathophysiological changes that may contribute to these disorders.
Collapse
Affiliation(s)
- Christopher Pittenger
- Departments of Psychiatry and Psychology, Yale Child Study Center, and Interdepartmental Neuroscience Program, Yale University School of Medicine, 34 Park Street, W315, New Haven, CT, 06519, USA.
| |
Collapse
|
17
|
Khoja S, Shah V, Garcia D, Asatryan L, Jakowec MW, Davies DL. Role of purinergic P2X4 receptors in regulating striatal dopamine homeostasis and dependent behaviors. J Neurochem 2016; 139:134-48. [PMID: 27402173 DOI: 10.1111/jnc.13734] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 06/26/2016] [Accepted: 06/28/2016] [Indexed: 11/27/2022]
Abstract
Purinergic P2X4 receptors (P2X4Rs) belong to the P2X superfamily of ion channels regulated by ATP. We recently demonstrated that P2X4R knockout (KO) mice exhibited deficits in sensorimotor gating, social interaction, and ethanol drinking behavior. Dopamine (DA) dysfunction may underlie these behavioral changes, but there is no direct evidence for P2X4Rs' role in DA neurotransmission. To test this hypothesis, we measured markers of DA function and dependent behaviors in P2X4R KO mice. P2X4R KO mice exhibited altered density of pre-synaptic markers including tyrosine hydroxylase, dopamine transporter; post-synaptic markers including dopamine receptors and phosphorylation of downstream targets including dopamine and cyclic-AMP regulated phosphoprotein of 32 kDa and cyclic-AMP-response element binding protein in different parts of the striatum. Ivermectin, an allosteric modulator of P2X4Rs, significantly affected dopamine and cyclic AMP regulated phosphoprotein of 32 kDa and extracellular regulated kinase1/2 phosphorylation in the striatum. Sensorimotor gating deficits in P2X4R KO mice were rescued by DA antagonists. Using the 6-hydroxydopamine model of DA depletion, P2X4R KO mice exhibited an attenuated levodopa (L-DOPA)-induced motor behavior, whereas ivermectin enhanced this behavior. Collectively, these findings identified an important role for P2X4Rs in maintaining DA homeostasis and illustrate how this association is important for CNS functions including motor control and sensorimotor gating. We propose that P2X4 receptors (P2X4Rs) regulate dopamine (DA) homeostasis and associated behaviors. Pre-synaptic and post-synaptic DA markers were significantly altered in the dorsal and ventral striatum of P2X4R KO mice, implicating altered DA neurotransmission. Sensorimotor gating deficits in P2X4R KO mice were rescued by DA antagonists. Ivermectin (IVM), a positive modulator of P2X4Rs, enhanced levodopa (L-DOPA)-induced motor behavior. These studies highlight potential interactions between P2X4Rs and DA system.
Collapse
Affiliation(s)
- Sheraz Khoja
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California, USA
| | - Vivek Shah
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Damaris Garcia
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Liana Asatryan
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, California, USA
| | - Michael W Jakowec
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Daryl L Davies
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
18
|
Methylone-induced hyperthermia and lethal toxicity: role of the dopamine and serotonin transporters. Behav Pharmacol 2016; 26:345-52. [PMID: 25794333 DOI: 10.1097/fbp.0000000000000135] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Methylone (2-methylamino-1-[3,4-methylenedioxy-phenyl]propan-1-one), an amphetamine analog, has emerged as a popular drug of abuse worldwide. Methylone induces hyperthermia, which is thought to contribute toward the lethal consequences of methylone overdose. Methylone has been assumed to induce hyperthermic effects through inhibition of serotonin and/or dopamine transporters (SERT and DAT, respectively). To examine the roles of each of these proteins in methylone-induced toxic effects, we used SERT and DAT knockout (KO) mice and assessed the hyperthermic and lethal effects caused by a single administration of methylone. Methylone produced higher rates of lethal toxicity compared with other amphetamine analogs in wild-type mice. Compared with wild-type mice, lethality was significantly lower in DAT KO mice, but not in SERT KO mice. By contrast, only a slight diminution in the hyperthermic effects of methylone was observed in DAT KO mice, whereas a slight enhancement of these effects was observed in SERT KO mice. Administration of the selective D1 receptor antagonist SCH 23390 and the D2 receptor antagonist raclopride reduced methylone-induced hyperthermia, but these drugs also had hypothermic effects in saline-treated mice, albeit to a smaller extent than the effects observed in methylone-treated mice. In contradistinction to 3,4-methylenedioxymethamphetamine, which induces its toxicity through SERT and DAT, these data indicate that DAT, but not SERT, is strongly associated with the lethal toxicity produced by methylone, which did not seem to be dependent on the hyperthermic effects of methylone. DAT is therefore a strong candidate molecule for interventions aimed at preventing acute neurotoxic and lethal effects of methylone.
Collapse
|
19
|
Lopes R, Soares R, Coelho R, Figueiredo-Braga M. Angiogenesis in the pathophysiology of schizophrenia — A comprehensive review and a conceptual hypothesis. Life Sci 2015; 128:79-93. [DOI: 10.1016/j.lfs.2015.02.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 01/27/2015] [Accepted: 02/12/2015] [Indexed: 01/11/2023]
|
20
|
John WS, Newman AH, Nader MA. Differential effects of the dopamine D3 receptor antagonist PG01037 on cocaine and methamphetamine self-administration in rhesus monkeys. Neuropharmacology 2015; 92:34-43. [PMID: 25576373 PMCID: PMC4346463 DOI: 10.1016/j.neuropharm.2014.12.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 12/17/2014] [Accepted: 12/22/2014] [Indexed: 12/17/2022]
Abstract
The dopamine D3 receptor (D3R) has been shown to mediate many of the behavioral effects of psychostimulants associated with high abuse potential. This study extended the assessment of the highly selective D3R antagonist PG01037 on cocaine and methamphetamine (MA) self-administration to include a food-drug choice procedure. Eight male rhesus monkeys (n=4/group) served as subjects in which complete cocaine and MA dose-response curves were determined daily in each session. When choice was stable, monkeys received acute and five-day treatment of PG01037 (1.0-5.6 mg/kg, i.v.). Acute administration of PG01037 was effective in reallocating choice from cocaine to food and decreasing cocaine intake, however, tolerance developed by day 5 of treatment. Up to doses that disrupted responding, MA choice and intake were not affected by PG01037 treatment. PG01037 decreased total reinforcers earned per session and the behavioral potency was significantly greater on MA-food choice compared to cocaine-food choice. Furthermore, the acute efficacy of PG01037 was correlated with the sensitivity of the D3/D2R agonist quinpirole to elicit yawning. These data suggest (1) that efficacy of D3R compounds in decreasing drug choice is greater in subjects with lower D3R, perhaps suggesting that it is percent occupancy that is the critical variable in determining efficacy and (2) differences in D3R activity in chronic cocaine vs. MA users. Although tolerance developed to the effects of PG01037 treatment on cocaine choice, tolerance did not develop to the disruptive effects on food-maintained responding. These findings suggest that combination treatments that decrease cocaine-induced elevations in DA may enhance the efficacy of D3R antagonists on cocaine self-administration.
Collapse
Affiliation(s)
- William S John
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, MD, USA.
| | - Michael A Nader
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
21
|
Hall FS, Sora I, Hen R, Uhl GR. Serotonin/dopamine interactions in a hyperactive mouse: reduced serotonin receptor 1B activity reverses effects of dopamine transporter knockout. PLoS One 2014; 9:e115009. [PMID: 25514162 PMCID: PMC4267809 DOI: 10.1371/journal.pone.0115009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 11/17/2014] [Indexed: 11/23/2022] Open
Abstract
Knockout (KO) mice that lack the dopamine transporter (SL6A3; DAT) display increased locomotion that can be attenuated, under some circumstances, by administration of drugs that normally produce psychostimulant-like effects, such as amphetamine and methylphenidate. These results have led to suggestions that DAT KO mice may model features of attention deficit hyperactivity disorder (ADHD) and that these drugs may act upon serotonin (5-HT) systems to produce these unusual locomotor decreasing effects. Evidence from patterns of brain expression and initial pharmacologic studies led us to use genetic and pharmacologic approaches to examine the influence of altered 5-HT1B receptor activity on hyperactivity in DAT KO mice. Heterozygous 5-HT1B KO and pharmacologic 5-HT1B antagonism both attenuated locomotor hyperactivity in DAT KO mice. Furthermore, DAT KO mice with reduced, but not eliminated, 5-HT1B receptor expression regained cocaine-stimulated locomotion, which was absent in DAT KO mice with normal levels of 5-HT1B receptor expression. Further experiments demonstrated that the degree of habituation to the testing apparatus determined whether cocaine had no effect on locomotion in DAT KO or reduced locomotion, helping to resolve differences among prior reports. These findings of complementation of the locomotor effects of DAT KO by reducing 5-HT1B receptor activity underscore roles for interactions between specific 5-HT receptors and dopamine (DA) systems in basal and cocaine-stimulated locomotion and support evaluation of 5-HT1B antagonists as potential, non-stimulant ADHD therapeutics.
Collapse
Affiliation(s)
- Frank Scott Hall
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, Ohio, United States of America
- Molecular Neurobiology Branch, National Institute on Drug Abuse – Intramural Research Program, Baltimore, Maryland, United States of America
- * E-mail:
| | - Ichiro Sora
- Kobe University Graduate School of Medicine, Kobe, Japan
| | - René Hen
- Departments of Pharmacology, Neuroscience and Pharmacology, Columbia University, New York, New York, United States of America; Division of Integrative Neuroscience, The New York State Psychiatric Institute, New York, New York, United States of America
| | - George R. Uhl
- Molecular Neurobiology Branch, National Institute on Drug Abuse – Intramural Research Program, Baltimore, Maryland, United States of America
| |
Collapse
|
22
|
Striatal dopamine receptor plasticity in neurotensin deficient mice. Behav Brain Res 2014; 280:160-71. [PMID: 25449842 DOI: 10.1016/j.bbr.2014.11.014] [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: 08/22/2014] [Revised: 11/04/2014] [Accepted: 11/07/2014] [Indexed: 11/21/2022]
Abstract
Schizophrenia is thought to be caused, at least in part, by dysfunction in striatal dopamine neurotransmission. Both clinical studies and animal research have implicated the dopamine neuromodulator neurotensin (NT) in the pathophysiology of schizophrenia. Utilizing male mice lacking the NT gene (NT(-/-)), these studies examined the consequences of NT deficiency on dopaminergic tone and function, investigating (1) dopamine concentrations and dopamine receptor and transporter expression and binding in dopaminergic terminal regions, and (2) the behavioral effects of selective dopamine receptor agonists on locomotion and sensorimotor gating in adult NT(-/-) mice compared to wildtype (NT(+/+)) mice. NT(-/-) mice did not differ from NT(+/+) mice in concentrations of dopamine or its metabolite DOPAC in any brain region examined. However, NT(-/-) mice showed significantly increased D1 receptor, D2 receptor, and dopamine transporter (DAT) mRNA in the caudate putamen compared to NT(+/+) controls. NT(-/-) mice also showed elevated D2 receptor binding densities in both the caudate putamen and nucleus accumbens shell compared to NT(+/+) mice. In addition, some of the behavioral effects of the D1-type receptor agonist SKF-82958 and the D2-type receptor agonist quinpirole on locomotion, startle amplitude, and prepulse inhibition were dose-dependently altered in NT(-/-) mice, showing altered D1-type and D2-type receptor sensitivity to stimulation by agonists in the absence of NT. The results indicate that NT deficiency alters striatal dopamine receptor expression, binding, and function. This suggests a critical role for the NT system in the maintenance of striatal DA system homeostasis and implicates NT deficiency in the etiology of dopamine-associated disorders such as schizophrenia.
Collapse
|
23
|
Nordenankar K, Smith-Anttila CJA, Schweizer N, Viereckel T, Birgner C, Mejia-Toiber J, Morales M, Leao RN, Wallén-Mackenzie Å. Increased hippocampal excitability and impaired spatial memory function in mice lacking VGLUT2 selectively in neurons defined by tyrosine hydroxylase promoter activity. Brain Struct Funct 2014; 220:2171-90. [PMID: 24802380 PMCID: PMC4481332 DOI: 10.1007/s00429-014-0778-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 04/11/2014] [Indexed: 02/01/2023]
Abstract
Three populations of neurons expressing the vesicular glutamate transporter 2 (Vglut2) were recently described in the A10 area of the mouse midbrain, of which two populations were shown to express the gene encoding, the rate-limiting enzyme for catecholamine synthesis, tyrosine hydroxylase (TH).One of these populations ("TH-Vglut2 Class1") also expressed the dopamine transporter (DAT) gene while one did not ("TH-Vglut2 Class2"), and the remaining population did not express TH at all ("Vglut2-only"). TH is known to be expressed by a promoter which shows two phases of activation, a transient one early during embryonal development, and a later one which gives rise to stable endogenous expression of the TH gene. The transient phase is, however, not specific to catecholaminergic neurons, a feature taken to advantage here as it enabled Vglut2 gene targeting within all three A10 populations expressing this gene, thus creating a new conditional knockout. These knockout mice showed impairment in spatial memory function. Electrophysiological analyses revealed a profound alteration of oscillatory activity in the CA3 region of the hippocampus. In addition to identifying a novel role for Vglut2 in hippocampus function, this study points to the need for improved genetic tools for targeting of the diversity of subpopulations of the A10 area.
Collapse
Affiliation(s)
- Karin Nordenankar
- Unit of Functional Neurobiology and Unit of Developmental Genetics, Biomedical Center, Department of Neuroscience, Uppsala University, Box 593, S-751 24, Uppsala, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Baldan LC, Williams KA, Gallezot JD, Pogorelov V, Rapanelli M, Crowley M, Anderson GM, Loring E, Gorczyca R, Billingslea E, Wasylink S, Panza KE, Ercan-Sencicek AG, Krusong K, Leventhal BL, Ohtsu H, Bloch MH, Hughes ZA, Krystal JH, Mayes L, de Araujo I, Ding YS, State MW, Pittenger C. Histidine decarboxylase deficiency causes tourette syndrome: parallel findings in humans and mice. Neuron 2014; 81:77-90. [PMID: 24411733 DOI: 10.1016/j.neuron.2013.10.052] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2013] [Indexed: 11/25/2022]
Abstract
Tourette syndrome (TS) is characterized by tics, sensorimotor gating deficiencies, and abnormalities of cortico-basal ganglia circuits. A mutation in histidine decarboxylase (Hdc), the key enzyme for the biosynthesis of histamine (HA), has been implicated as a rare genetic cause. Hdc knockout mice exhibited potentiated tic-like stereotypies, recapitulating core phenomenology of TS; these were mitigated by the dopamine (DA) D2 antagonist haloperidol, a proven pharmacotherapy, and by HA infusion into the brain. Prepulse inhibition was impaired in both mice and humans carrying Hdc mutations. HA infusion reduced striatal DA levels; in Hdc knockout mice, striatal DA was increased and the DA-regulated immediate early gene Fos was upregulated. DA D2/D3 receptor binding was altered both in mice and in humans carrying the Hdc mutation. These data confirm histidine decarboxylase deficiency as a rare cause of TS and identify HA-DA interactions in the basal ganglia as an important locus of pathology.
Collapse
Affiliation(s)
| | - Kyle A Williams
- Department of Psychiatry, Yale University School of Medicine.,Department of Child Study Center, Yale University School of Medicine
| | | | | | | | - Michael Crowley
- Department of Child Study Center, Yale University School of Medicine
| | - George M Anderson
- Department of Child Study Center, Yale University School of Medicine.,Department of Laboratory Medicine, Yale University School of Medicine
| | - Erin Loring
- Department of Child Study Center, Yale University School of Medicine.,Department of Genetics, Yale University School of Medicine.,Department of Program on Neurogenetics, Yale University School of Medicine
| | | | | | | | - Kaitlyn E Panza
- Department of Child Study Center, Yale University School of Medicine
| | - A Gulhan Ercan-Sencicek
- Department of Child Study Center, Yale University School of Medicine.,Department of Genetics, Yale University School of Medicine
| | - Kuakarun Krusong
- Department of Psychiatry, Yale University School of Medicine.,Dept. of Biochem., Faculty of Science, Chulalongkorn Univ., Bangkok, Thailand
| | - Bennett L Leventhal
- Nathan S. Kline Institute for Psychiatric Research.,New York University Dept of Child and Adolescent Psychiatry
| | - Hiroshi Ohtsu
- Tohoku University, Graduate School of Engineering, Sendai, Japan
| | - Michael H Bloch
- Department of Psychiatry, Yale University School of Medicine.,Department of Child Study Center, Yale University School of Medicine
| | - Zoë A Hughes
- Neuroscience Research Unit, Pfizer, Inc., Cambridge, MA
| | - John H Krystal
- Department of Psychiatry, Yale University School of Medicine
| | - Linda Mayes
- Department of Psychiatry, Yale University School of Medicine.,Department of Child Study Center, Yale University School of Medicine.,Department of Pediatrics, Yale University School of Medicine.,Department of Psychology, Yale University School of Medicine
| | - Ivan de Araujo
- Department of Psychiatry, Yale University School of Medicine.,John B. Pierce Laboratory, New Haven, CT
| | - Yu-Shin Ding
- Department of Diagnostic Radiology, Yale University School of Medicine
| | - Matthew W State
- Department of Psychiatry, Yale University School of Medicine.,Department of Child Study Center, Yale University School of Medicine.,Department of Genetics, Yale University School of Medicine.,Department of Program on Neurogenetics, Yale University School of Medicine
| | - Christopher Pittenger
- Department of Psychiatry, Yale University School of Medicine.,Department of Child Study Center, Yale University School of Medicine.,Department of Psychology, Yale University School of Medicine.,Integrated Neuroscience Research Program; New Haven, CT 06520
| |
Collapse
|
25
|
Lopes R, Soares R, Figueiredo-Braga M, Coelho R. Schizophrenia and cancer: is angiogenesis a missed link? Life Sci 2014; 97:91-5. [PMID: 24378672 DOI: 10.1016/j.lfs.2013.12.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 12/08/2013] [Accepted: 12/16/2013] [Indexed: 01/11/2023]
Abstract
Cancer prevalence and risk in schizophrenia (SZ) patients, as well as their implicated molecular pathways, is a debate that has become increasingly appreciated, despite lacking evidence. Since angiogenesis is imbalanced in both conditions, a non-systematic review of the existing literature using the PubMed database was performed to summarize current knowledge and to elucidate hypothesis regarding the reduced incidence of cancer in SZ, exploring possible angiogenesis biology aspects that can be interrelated both with SZ and cancer. Some lines of evidence based in epidemiology, genetic, molecular and biochemical studies suggest a putative interplay between SZ pathophysiology and angiogenesis, involving different molecular pathways and also influencing cancer biology. Studying angiogenesis in SZ and its implications to cancer is an unexplored field that could provide more insightful knowledge regarding its pathophysiology and promote the development of treatment applications.
Collapse
Affiliation(s)
- Rui Lopes
- Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; Clinic of Psychiatry and Mental Health, Centro Hospitalar de São João, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal.
| | - Raquel Soares
- Department of Biochemistry (U38-FCT), Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal.
| | - Margarida Figueiredo-Braga
- Department of Clinical Neurosciences and Mental Health, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal.
| | - Rui Coelho
- Clinic of Psychiatry and Mental Health, Centro Hospitalar de São João, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; Department of Clinical Neurosciences and Mental Health, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal.
| |
Collapse
|
26
|
Wood S, Sage JR, Shuman T, Anagnostaras SG. Psychostimulants and cognition: a continuum of behavioral and cognitive activation. Pharmacol Rev 2013; 66:193-221. [PMID: 24344115 PMCID: PMC3880463 DOI: 10.1124/pr.112.007054] [Citation(s) in RCA: 169] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Psychostimulants such as cocaine have been used as performance enhancers throughout recorded history. Although psychostimulants are commonly prescribed to improve attention and cognition, a great deal of literature has described their ability to induce cognitive deficits, as well as addiction. How can a single drug class be known to produce both cognitive enhancement and impairment? Properties of the particular stimulant drug itself and individual differences between users have both been suggested to dictate the outcome of stimulant use. A more parsimonious alternative, which we endorse, is that dose is the critical determining factor in cognitive effects of stimulant drugs. Herein, we review several popular stimulants (cocaine, amphetamine, methylphenidate, modafinil, and caffeine), outlining their history of use, mechanism of action, and use and abuse today. One common graphic depiction of the cognitive effects of psychostimulants is an inverted U-shaped dose-effect curve. Moderate arousal is beneficial to cognition, whereas too much activation leads to cognitive impairment. In parallel to this schematic, we propose a continuum of psychostimulant activation that covers the transition from one drug effect to another as stimulant intake is increased. Low doses of stimulants effect increased arousal, attention, and cognitive enhancement; moderate doses can lead to feelings of euphoria and power, as well as addiction and cognitive impairment; and very high doses lead to psychosis and circulatory collapse. This continuum helps account for the seemingly disparate effects of stimulant drugs, with the same drug being associated with cognitive enhancement and impairment.
Collapse
Affiliation(s)
- Suzanne Wood
- 9500 Gilman Dr MC 0109, La Jolla, CA 92093-0109.
| | | | | | | |
Collapse
|
27
|
Mereu M, Bonci A, Newman AH, Tanda G. The neurobiology of modafinil as an enhancer of cognitive performance and a potential treatment for substance use disorders. Psychopharmacology (Berl) 2013; 229:415-34. [PMID: 23934211 PMCID: PMC3800148 DOI: 10.1007/s00213-013-3232-4] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 07/28/2013] [Indexed: 12/31/2022]
Abstract
RATIONALE AND OBJECTIVES Modafinil (MOD) and its R-enantiomer (R-MOD) are approved medications for narcolepsy and other sleep disorders. They have also been used, off-label, as cognitive enhancers in populations of patients with mental disorders, including substance abusers that demonstrate impaired cognitive function. A debated nonmedical use of MOD in healthy individuals to improve intellectual performance is raising questions about its potential abuse liability in this population. RESULTS AND CONCLUSIONS MOD has low micromolar affinity for the dopamine transporter (DAT). Inhibition of dopamine (DA) reuptake via the DAT explains the enhancement of DA levels in several brain areas, an effect shared with psychostimulants like cocaine, methylphenidate, and the amphetamines. However, its neurochemical effects and anatomical pattern of brain area activation differ from typical psychostimulants and are consistent with its beneficial effects on cognitive performance processes such as attention, learning, and memory. At variance with typical psychostimulants, MOD shows very low, if any, abuse liability, in spite of its use as a cognitive enhancer by otherwise healthy individuals. Finally, recent clinical studies have focused on the potential use of MOD as a medication for treatment of drug abuse, but have not shown consistent outcomes. However, positive trends in several result measures suggest that medications that improve cognitive function, like MOD or R-MOD, may be beneficial for the treatment of substance use disorders in certain patient populations.
Collapse
Affiliation(s)
- Maddalena Mereu
- Molecular Targets & Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, DHHS; 251 Bayview Blvd., NIDA suite 200, Baltimore, MD, 21224
| | - Antonello Bonci
- Synaptic Plasticity Section, Cellular Neurobiology Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, DHHS; 251 Bayview Blvd., NIDA suite 200, Baltimore, MD, 21224
| | - Amy Hauck Newman
- Molecular Targets & Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, DHHS; 251 Bayview Blvd., NIDA suite 200, Baltimore, MD, 21224
| | - Gianluigi Tanda
- Molecular Targets & Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, DHHS; 251 Bayview Blvd., NIDA suite 200, Baltimore, MD, 21224
| |
Collapse
|
28
|
Lee MY, Yu JH, Kim JY, Seo JH, Park ES, Kim CH, Kim H, Cho SR. Alteration of synaptic activity-regulating genes underlying functional improvement by long-term exposure to an enriched environment in the adult brain. Neurorehabil Neural Repair 2013; 27:561-74. [PMID: 23558143 DOI: 10.1177/1545968313481277] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Housing animals in an enriched environment (EE) enhances behavioral function. However, the mechanism underlying this EE-mediated functional improvement and the resultant changes in gene expression have yet to be elucidated. OBJECTIVES We attempted to investigate the underlying mechanisms associated with long-term exposure to an EE by evaluating gene expression patterns. METHODS We housed 6-week-old CD-1 (ICR) mice in standard cages or an EE comprising a running wheel, novel objects, and social interaction for 2 months. Motor and cognitive performances were evaluated using the rotarod test and passive avoidance test, and gene expression profile was investigated in the cerebral hemispheres using microarray and gene set enrichment analysis (GSEA). RESULTS In behavioral assessment, an EE significantly enhanced rotarod performance and short-term working memory. Microarray analysis revealed that genes associated with neuronal activity were significantly altered by an EE. GSEA showed that genes involved in synaptic transmission and postsynaptic signal transduction were globally upregulated, whereas those associated with reuptake by presynaptic neurotransmitter transporters were downregulated. In particular, both microarray and GSEA demonstrated that EE exposure increased opioid signaling, acetylcholine release cycle, and postsynaptic neurotransmitter receptors but decreased Na+ / Cl- -dependent neurotransmitter transporters, including dopamine transporter Slc6a3 in the brain. Western blotting confirmed that SLC6A3, DARPP32 (PPP1R1B), and P2RY12 were largely altered in a region-specific manner. CONCLUSION An EE enhanced motor and cognitive function through the alteration of synaptic activity-regulating genes, improving the efficient use of neurotransmitters and synaptic plasticity by the upregulation of genes associated with postsynaptic receptor activity and downregulation of presynaptic reuptake by neurotransmitter transporters.
Collapse
Affiliation(s)
- Min-Young Lee
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
29
|
O'Connell LA, Mitchell MM, Hofmann HA, Crews D. Androgens coordinate neurotransmitter-related gene expression in male whiptail lizards. GENES BRAIN AND BEHAVIOR 2012; 11:813-8. [PMID: 22862958 DOI: 10.1111/j.1601-183x.2012.00828.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 06/24/2012] [Accepted: 07/20/2012] [Indexed: 11/29/2022]
Abstract
Sex steroid hormones coordinate neurotransmitter systems in the male brain to facilitate sexual behavior. Although neurotransmitter release in the male brain has been well documented, little is known about how androgens orchestrate changes in gene expression of neurotransmitter receptors. We used male whiptail lizards (Cnemidophorus inornatus) to investigate how androgens alter neurotransmitter-related gene expression in brain regions involved in social decision making. We focused on three neurotransmitter systems involved in male-typical sexual behavior, including the N-methyl-d-aspartate (NMDA) glutamate receptor, nitric oxide and dopamine receptors. Here, we show that in androgen-treated males, there are coordinated changes in neurotransmitter-related gene expression. In androgen-implanted castrates compared with blank-implanted castrates (control group), we found associated increases in neuronal nitric oxide synthase gene expression in the nucleus accumbens (NAcc), preoptic area and ventromedial hypothalamus, a decrease of NR1 gene expression (obligate subunit of NMDA receptors) in the medial amygdaloid area and NAcc and a decrease in D1 and D2 dopamine receptor gene expression in the NAcc. Our results support and expand the current model of androgen-mediated gene expression changes of neurotransmitter-related systems that facilitate sexual behavior in males. This also suggests that the proposed evolutionarily ancient reward system that reinforces sexual behavior in amniote vertebrates extends to reptiles.
Collapse
Affiliation(s)
- L A O'Connell
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA
| | | | | | | |
Collapse
|
30
|
Ramadan E, Chang L, Chen M, Ma K, Hall FS, Uhl GR, Rapoport SI, Basselin M. Knocking out the dopamine reuptake transporter (DAT) does not change the baseline brain arachidonic acid signal in the mouse. Int J Neurosci 2012; 122:373-80. [PMID: 22376027 PMCID: PMC3464054 DOI: 10.3109/00207454.2012.665972] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Dopamine transporter (DAT) homozygous knockout (DAT(-/-)) mice have a 10-fold higher extracellular (DA) concentration in the caudate-putamen and nucleus accumbens than do wildtype (DAT(+/+)) mice, but show reduced presynaptic DA synthesis and fewer postsynaptic D(2) receptors. One aspect of neurotransmission involves DA binding to postsynaptic D(2)-like receptors coupled to cytosolic phospholipase A(2) (cPLA(2)), which releases the second messenger, arachidonic acid (AA), from synaptic membrane phospholipid. We hypothesized that tonic overactivation of D(2)-like receptors in DAT(-/-) mice due to the excess DA would not increase brain AA signaling, because of compensatory downregulation of postsynaptic DA signaling mechanisms. METHODS [1-(14)C]AA was infused intravenously for 3 min in unanesthetized DAT(+/+), heterozygous (DAT(+/-)), and DAT(-/-) mice. AA incorporation coefficients k* and rates J(in), markers of AA metabolism and signaling, were imaged in 83 brain regions using quantitative autoradiography; brain cPLA(2)-IV activity also was measured. RESULTS Neither k* nor J(in) for AA in any brain region, or brain cPLA(2)-IV activity, differed significantly among DAT(-/-), DAT(+/-), and DAT(+/+) mice. CONCLUSIONS These results differ from reported increases in k* and J(in) for AA, and in brain cPLA(2) expression, in serotonin reuptake transporter (5-HTT) knockout mice, and suggest that postsynaptic dopaminergic neurotransmission mechanisms involving AA are downregulated despite elevated DA in DAT(-/-) mice.
Collapse
Affiliation(s)
- Epolia Ramadan
- Brain Physiology and Metabolism Section, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Meck WH, Cheng RK, MacDonald CJ, Gainetdinov RR, Caron MG, Çevik MÖ. Gene-dose dependent effects of methamphetamine on interval timing in dopamine-transporter knockout mice. Neuropharmacology 2012; 62:1221-9. [DOI: 10.1016/j.neuropharm.2011.01.042] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 01/19/2011] [Accepted: 01/24/2011] [Indexed: 10/18/2022]
|
32
|
Abstract
The neonatal 6-OHDA-lesioned rat, coloboma mouse, DAT-KO mouse, and spontaneously hypertensive rat (SHR) models all bear a phenotypic resemblance to ADHD in that they express hyperactivity, inattention, and/or impulsivity. The models also illustrate the heterogeneity of ADHD: the initial cause (chemical depletion or genetic abnormality) of the ADHD-like behaviors is different for each model. Neurochemical and behavioral studies of the models indicate aberrations in monoaminergic neurotransmission. Hyperdopaminergic neurotransmission is implicated in the abnormal behavior of all models. Norepinephrine has a dual enhancing/inhibitory role in ADHD symptoms, and serotonin acts to inhibit abnormal dopamine and norepinephrine signaling. It is unlikely that symptoms arise from a single neurotransmitter dysfunction. Rather, studies of animal models of ADHD suggest that symptoms develop through the complex interactions of monoaminergic neurotransmitter systems.
Collapse
Affiliation(s)
- Xueliang Fan
- Departments of Pharmacology and Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | | | | |
Collapse
|
33
|
Abstract
Modafinil (2-((diphenylmethyl)sulfinyl)acetamide) is described as an atypical stimulant and is a putative cognition enhancer for schizophrenia, but the precise mechanisms of action remain unclear. Receptor knockout (KO) mice offer an opportunity to identify receptors that contribute to a drug-induced effect. Here we examined the effects of modafinil on exploration in C57BL/6J mice, in dopamine drd1, drd2, drd3, and drd4 wild-type (WT), heterozygous (HT), and KO mice, and in 129/SJ mice pretreated with the drd1 antagonist SCH23390 using a cross-species test paradigm based on the behavioral pattern monitor. Modafinil increased activity, specific exploration (rearing), and the smoothness of locomotor paths (reduced spatial d) in C57BL/6J and 129/SJ mice (increased holepoking was also observed in these mice). These behavioral profiles are similar to that produced by the dopamine transporter inhibitor GBR12909. Modafinil was ineffective at increasing activity in male drd1 KOs, rearing in female drd1 KOs, or reducing spatial d in all drd1 KOs, but produced similar effects in drd1 WT and HT mice as in C57BL/6J mice. Neither dopamine drd2 nor drd3 mutants attenuated modafinil-induced effects. Drd4 mutants exhibited a genotype dose-dependent attenuation of modafinil-induced increases in specific exploration. Furthermore, the drd1 KO effects were largely supported by the SCH23390 study. Thus, the dopamine drd1 receptor appears to exert a primary role in modafinil-induced effects on spontaneous exploration, whereas the dopamine drd4 receptor appears to be important for specific exploration. The modafinil-induced alterations in exploratory behavior may reflect increased synaptic dopamine and secondary actions mediated by dopamine drd1 and drd4 receptors.
Collapse
|
34
|
Berlanga ML, Price DL, Phung BS, Giuly R, Terada M, Yamada N, Cyr M, Caron MG, Laakso A, Martone ME, Ellisman MH. Multiscale imaging characterization of dopamine transporter knockout mice reveals regional alterations in spine density of medium spiny neurons. Brain Res 2011; 1390:41-9. [PMID: 21439946 DOI: 10.1016/j.brainres.2011.03.044] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 03/16/2011] [Accepted: 03/17/2011] [Indexed: 11/26/2022]
Abstract
The dopamine transporter knockout (DAT KO) mouse is a model of chronic hyperdopaminergia used to study a wide range of neuropsychiatric disorders such as schizophrenia, attention deficit hyperactivity disorder (ADHD), drug abuse, depression, and Parkinson's disease (PD). Early studies characterizing this mouse model revealed a subtle, but significant, decrease in the anterior striatal volume of DAT KO mice accompanied by a decrease in neuronal cell body numbers (Cyr et al., 2005). The present studies were conducted to examine medium spiny neuron (MSN) morphology by extending these earlier reports to include multiscale imaging studies using correlated light microscopy (LM) and electron microscopy (EM) techniques. Specifically, we set out to determine if chronic hyperdopaminergia results in quantifiable or qualitative changes in DAT KO mouse MSNs relative to wild-type (WT) littermates. Using Neurolucida Explorer's morphometric analysis, we measured spine density, dendritic length and synapse number at ages that correspond with the previously reported changes in striatal volume and progressive cell loss. Light microscopic analysis using Neurolucida tracings of photoconverted striatal MSNs revealed a highly localized loss of dendritic spines on the proximal portion of the dendrite (30 μm from the soma) in the DAT KO group. Next, thick sections containing MSN dendritic segments located at a distance of 20-60 μm from the cell soma, a region of the dendrite where spine density is reported to be the highest, were analyzed using electron microscope tomography (EMT). Because of the resolution limits of LM, the EM analysis was an extra measure taken to assure that our analysis included nearly all spines. Spine density measurements collected from the EMT data revealed only a modest decrease in the DAT KO group (n=3 mice) compared to age-matched WT controls (n=3 mice), a trend that supports the LM findings. Finally, a synaptic quantification using unbiased stereology did not detect a difference between DAT KO mice (n=6 mice) and WT controls (n=7 mice) at the EM level, supporting the focal nature of the early synaptic loss. These findings suggest that DAT KO mice have MSNs with highly localized spine loss and not an overall morphologically distinct cell shape. The characterization of morphological changes in DAT KO mice may provide information about the neural substrates underlying altered behaviors in these mice, with relevance for human neurological disorders thought to involve altered dopaminergic homeostasis. Results from this study also indicate the difficulty in correlating structural changes across scales, as the results on fine structure revealed thus far are subtle and non-uniform across striatal MSNs. The complexities associated with multiscale studies are driving the development of shared online informatics resources by gaining access to data where it is being analyzed.
Collapse
Affiliation(s)
- M L Berlanga
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California, San Diego, La Jolla, CA 92093-0608, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Haenisch B, Bönisch H. Depression and antidepressants: Insights from knockout of dopamine, serotonin or noradrenaline re-uptake transporters. Pharmacol Ther 2011; 129:352-68. [DOI: 10.1016/j.pharmthera.2010.12.002] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Accepted: 11/29/2010] [Indexed: 12/15/2022]
|
36
|
Kanno K, Ishiura S. Differential effects of the HESR/HEY transcription factor family on dopamine transporter reporter gene expression via variable number of tandem repeats. J Neurosci Res 2011; 89:562-75. [DOI: 10.1002/jnr.22593] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 11/10/2010] [Accepted: 12/09/2010] [Indexed: 11/08/2022]
|
37
|
Teixeira A, Müller L, Reckziegel P, Boufleur N, Pase C, Villarinho J, Fachinetto R, Ferreira J, Rocha J, Bürger M. Beneficial effects of an innovative exercise model on motor and oxidative disorders induced by haloperidol in rats. Neuropharmacology 2011; 60:432-8. [DOI: 10.1016/j.neuropharm.2010.10.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 09/27/2010] [Accepted: 10/20/2010] [Indexed: 10/18/2022]
|
38
|
Ramamoorthy S, Shippenberg TS, Jayanthi LD. Regulation of monoamine transporters: Role of transporter phosphorylation. Pharmacol Ther 2010; 129:220-38. [PMID: 20951731 DOI: 10.1016/j.pharmthera.2010.09.009] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Accepted: 09/24/2010] [Indexed: 10/18/2022]
Abstract
Presynaptic biogenic amine transporters mediate reuptake of released amines from the synapse, thus regulating serotonin, dopamine and norepinephrine neurotransmission. Medications utilized in the treatment of depression, attention deficit-hyperactivity disorder and other psychiatric disorders possess high affinity for amine transporters. In addition, amine transporters are targets for psychostimulants. Altered expression of biogenic amine transporters has long been implicated in several psychiatric and degenerative disorders. Therefore, appropriate regulation and maintenance of biogenic amine transporter activity is critical for the maintenance of normal amine homoeostasis. Accumulating evidence suggests that cellular protein kinases and phosphatases regulate amine transporter expression, activity, trafficking and degradation. Amine transporters are phosphoproteins that undergo dynamic control under the influence of various kinase and phosphatase activities. This review presents a brief overview of the role of amine transporter phosphorylation in the regulation of amine transport in the normal and diseased brain. Understanding the molecular mechanisms by which phosphorylation events affect amine transporter activity is essential for understanding the contribution of transporter phosphorylation to the regulation of monoamine neurotransmission and for identifying potential new targets for the treatment of various brain diseases.
Collapse
Affiliation(s)
- Sammanda Ramamoorthy
- Department of Neurosciences, Division of Neuroscience Research, Medical University of South Carolina, Charleston, South Carolina 29425, USA.
| | | | | |
Collapse
|
39
|
Hahn T, Heinzel S, Dresler T, Plichta MM, Renner TJ, Markulin F, Jakob PM, Lesch KP, Fallgatter AJ. Association between reward-related activation in the ventral striatum and trait reward sensitivity is moderated by dopamine transporter genotype. Hum Brain Mapp 2010; 32:1557-65. [PMID: 20845394 DOI: 10.1002/hbm.21127] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Revised: 05/14/2010] [Accepted: 06/21/2010] [Indexed: 12/31/2022] Open
Abstract
The impact of individual differences on human reward processing has been a focus of research in recent years, particularly, as they are associated with a variety of neuropsychiatric diseases including addiction and attention-deficit/hyperactivity disorder. Studies exploring the neural basis of individual differences in reward sensitivity have consistently implicated the ventral striatum (VS) as a core component of the human reward system. However, the mechanisms of dopaminergic neurotransmission underlying ventral striatal activation as well as trait reward sensitivity remain speculative. We addressed this issue by investigating the triadic interplay between VS reactivity during reward anticipation using functional magnetic resonance imaging, trait reward sensitivity, and dopamine (DA) transporter genotype (40-bp 3'VNTR of DAT, SLC6A3) affecting synaptic DA neurotransmission. Our results show that DAT variation moderates the association between VS-reactivity and trait reward sensitivity. Specifically, homozygote carriers of the DAT 10-repeat allele exhibit a strong positive correlation between reward sensitivity and reward-related VS activity whereas this relationship is absent in the DAT 9-repeat allele carriers. We discuss the possibility that this moderation of VS-trait relation might arise from DAT-dependent differences in DA availability affecting synaptic plasticity within the VS. Generally, studying the impact of dopaminergic gene variations on the relation between reward-related brain activity and trait reward sensitivity might facilitate the investigation of complex mechanisms underlying disorders linked to dysregulation of DA neurotransmission.
Collapse
Affiliation(s)
- Tim Hahn
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, 97080 Würzburg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Xu J, Hassanzadeh B, Chu W, Tu Z, Jones LA, Luedtke RR, Perlmutter JS, Mintun MA, Mach RH. [3H]4-(dimethylamino)-N-(4-(4-(2-methoxyphenyl)piperazin-1-yl) butyl)benzamide: a selective radioligand for dopamine D(3) receptors. II. Quantitative analysis of dopamine D(3) and D(2) receptor density ratio in the caudate-putamen. Synapse 2010; 64:449-59. [PMID: 20175227 DOI: 10.1002/syn.20748] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
4-(Dimethylamino)-N-(4-(4-(2-methoxyphenyl)piperazin-1-yl)butyl)benzamide (WC-10), a N-phenyl piperazine analog, displays high affinity and moderate selectivity for dopamine D(3) receptors versus dopamine D(2) receptors (Chu et al. [2005] Bioorg Med Chem 13:77-87). In this study, WC-10 was radiolabeled with tritium (specific activity = 80 Ci/mmol), and quantitative autoradiography studies were conducted using rhesus monkey and Sprague-Dawley rat brain sections. K(d) values for the binding of [3H]WC-10 to D(3) receptors obtained from quantitative autoradiography with rhesus monkey and rat brain sections are in agreement with K(d) values obtained from cloned human and rat receptors (Xu et al. [2009] Synapse 63:717-728). The D(2) selective antagonist [3H]raclopride binds with 11-fold higher affinity to human HEK D(2L) (K(d) = 1.6 nM) than HEK D(3) (K(d) = 18 nM) receptors; [3H]raclopride binds to rat Sf9 rD(2L) receptors with a K(d) of 6.79 nM, a value that is 4-fold lower than binding to human HEK D(2L) receptors and 2.5-fold higher than binding to rat Sf9 rD(3) receptors. In vitro quantitative autoradiography studies with [3H]WC-10 and [3H]raclopride were conducted on adult rat and rhesus monkey brain sections. A mathematical model for calculating the absolute densities of dopamine D(2) and D(3) receptors based on the in vitro receptor binding data of [3H]WC-10 and [3H]raclopride was developed.
Collapse
Affiliation(s)
- Jinbin Xu
- Department of Radiology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Guillin O, Abi-Dargham A, Laruelle M. Neurobiology of dopamine in schizophrenia. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2009; 78:1-39. [PMID: 17349856 DOI: 10.1016/s0074-7742(06)78001-1] [Citation(s) in RCA: 209] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This chapter is an update on the dopamine (DA) imbalance in schizophrenia, including the evidence for subcortical hyperstimulation of D2 receptors underlying positive symptoms and cortical hypodopaminergia-mediating cognitive disturbances and negative symptoms. After a brief review of the anatomical neurocircuitry of this transmitter system as a background, we summarize the evidence for dopaminergic alterations deriving from pharmacological, postmortem, and imaging studies. This evidence supports a prominent role for D2 antagonism in the treatment of positive symptoms of schizophrenia and strongly suggests the need for alternative approaches to address the more challenging problem of negative symptoms and cognitive disturbances.
Collapse
Affiliation(s)
- Olivier Guillin
- Department of Psychiatry, Columbia College of Physicians and Surgeons, New York State Psychiatric Institute, Columbia University, New York 10032, USA
| | | | | |
Collapse
|
42
|
Xu J, Chu W, Tu Z, Jones LA, Luedtke RR, Perlmutter JS, Mintun MA, Mach RH. [(3)H]4-(Dimethylamino)-N-[4-(4-(2-methoxyphenyl)piperazin- 1-yl)butyl]benzamide, a selective radioligand for dopamine D(3) receptors. I. In vitro characterization. Synapse 2009; 63:717-28. [PMID: 19425052 DOI: 10.1002/syn.20652] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
4-(Dimethylamino)-N-(4-(4-(2-methoxyphenyl)piperazin-1-yl)butyl)benzamide (WC-10), a N-phenyl piperazine analog, has been shown to have high affinity and selectivity for dopamine D(3) receptors versus dopamine D(2) receptors (Chu et al. [2005] Bioorg Med Chem 13:77-87). In this study, WC-10 was radiolabeled with tritium (specific activity = 80 Ci/mmol) and [(3)H]WC-10 binding to genetically cloned dopamine D(2L) and D(3) receptors was evaluated in vitro. [(3)H]WC-10 binds with a 66-fold higher affinity to human HEK D(3) than HEK D(2L) receptors, with a dissociation constant (K(d)) of 1.2 nM at HEK D(3) receptors. However, [(3)H]WC-10 binds to rat Sf9 rD(3) receptors with a K(d) of 3.9 nM, a value that is 3-fold lower than binding to human HEK D(3) receptors and 40-fold value higher than binding to rat Sf9 rD(2L) receptors. The K(d) values obtained from saturation binding experiments were consistent with the results determined from kinetic (k(on) and k(off)) studies. The pharmacologic profiles of a series of dopaminergic drugs for inhibiting the binding of [(3)H]WC-10 to D(3) receptors was in agreement with previously reported data. In vitro autoradiography studies of rat and monkey brains show that [(3)H]WC-10 labeled D(3) sites in the striatal region.
Collapse
Affiliation(s)
- Jinbin Xu
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Analysis of motor function and dopamine systems of SAMP6 mouse. Physiol Behav 2009; 96:464-9. [DOI: 10.1016/j.physbeh.2008.11.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Revised: 10/16/2008] [Accepted: 11/21/2008] [Indexed: 11/23/2022]
|
44
|
Ghisi V, Ramsey AJ, Masri B, Gainetdinov RR, Caron MG, Salahpour A. Reduced D2-mediated signaling activity and trans-synaptic upregulation of D1 and D2 dopamine receptors in mice overexpressing the dopamine transporter. Cell Signal 2008; 21:87-94. [PMID: 18929645 DOI: 10.1016/j.cellsig.2008.09.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Accepted: 09/26/2008] [Indexed: 11/17/2022]
Abstract
The dopamine transporter (DAT) regulates the temporal and spatial actions of dopamine by reuptaking this neurotransmitter into the presynaptic neurons. We recently generated transgenic mice overexpressing DAT (DAT-tg) that have a 3-fold increase in DAT protein levels which results in a 40% reduction of the extracellular DA concentration in the striatum. The aim of this study was to examine the effect of this reduction in dopaminergic tone on postsynaptic responses mediated by dopamine receptors. We report here that DAT-tg mice have increased levels of striatal D1 (30%) and D2 (approximately 60%) dopamine receptors with D1 receptor signaling components not significantly altered, as evidenced by unaffected basal or stimulated levels of phospho-GluR1 (Ser845) and phospho-ERK2. However, the novel D2 mediated Akt signaling is markedly altered in DAT-tg animals. In particular, there is a 300% increase in the basal levels of phospho-Akt in the striatum of DAT-tg, reflecting the reduced extracellular dopamine tone in these animals. This increase in basal pAkt levels can be pharmacologically recapitulated by partial dopamine depletion in WT mice treated with the selective tyrosine hydroxylase inhibitor alpha-methyl-para-tyrosine (alpha-MPT). Behaviorally, DAT-tg animals demonstrate an augmented synergistic interaction between up-regulated D1 and D2 receptors, which results in increased climbing behavior in transgenic mice after stimulation with either apomorphine or a co-administration of selective D1 and D2 receptor agonists. In sum, our study reveals that hypodopaminegia caused by up-regulation of DAT results in significant alterations at postsynaptic receptor function with most notable dysregulation at the level of D2 receptor signaling.
Collapse
Affiliation(s)
- Valentina Ghisi
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
Modafinil is a wake-promoting compound with low abuse potential used in the treatment of narcolepsy. Although the compound is reported to affect multiple neurotransmitter systems such as catecholamines, serotonin, glutamate, GABA, orexin, and histamine, however, the molecular mechanism by which modafinil increases wakefulness is debated. Herein we used dopamine (DA) D(2) receptor (D(2)R)-deficient mice combined with D(1)R- and D(2)R-specific antagonists to clarify the role of DA receptors in the arousal effects of modafinil. In wild-type mice, intraperitoneal modafinil induced wakefulness in a dose-dependent manner. Pretreatment with either D(1)R antagonist SCH23390 [R-(+)-7-chloro-8-hydroxy-3-methyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepine] at 30 microg/kg or D(2)R antagonist raclopride at 2 mg/kg blocked the arousal effects of low-dose modafinil at 22.5 and 45 mg/kg. When modafinil was given at 90 and 180 mg/kg, pretreatment of D(1)R antagonist did not affect the wakefulness at all, whereas D(2)R antagonist significantly attenuated the wakefulness to the half level compared with vehicle control. Similarly, D(2)R knock-out (KO) mice exhibited attenuated modafinil-induced wakefulness. However, pretreatment of D(2)R KO mice with D(1)R antagonist completely abolished arousal effects of modafinil. These findings strongly indicate that dopaminergic D(1)R and D(2)R are essential for the wakefulness induced by modafinil.
Collapse
|
46
|
Sevak RJ, Koek W, Owens WA, Galli A, Daws LC, France CP. Feeding conditions differentially affect the neurochemical and behavioral effects of dopaminergic drugs in male rats. Eur J Pharmacol 2008; 592:109-15. [PMID: 18652823 DOI: 10.1016/j.ejphar.2008.07.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2007] [Revised: 06/24/2008] [Accepted: 07/05/2008] [Indexed: 10/21/2022]
Abstract
The high co-morbidity of eating disorders and substance abuse suggests that nutritional status can impact vulnerability to drug abuse. These studies used rats to examine the effects of food restriction on dopamine clearance in striatum and on the behavioral effects of amphetamine (locomotion, conditioned place preference), the dopamine receptor agonist quinpirole (yawning), and the dopamine receptor antagonist raclopride (catalepsy). Amphetamine increased locomotion and produced conditioned place preference. Food restriction reduced dopamine clearance, which was restored by repeated treatment with amphetamine or by free feeding. Food restriction also decreased sensitivity to quinpirole-induced yawning and raclopride-induced catalepsy; normal sensitivity to both drugs was restored by free feeding. The same amphetamine treatment that normalized dopamine clearance, failed to restore normal sensitivity to quinpirole or raclopride, suggesting that in food-restricted rats the activity of dopamine transporters and dopamine receptors is differentially affected by pathways that are stimulated by amphetamine. These studies show that modest changes in nutritional status markedly alter dopamine neurotransmission and the behavioral effects of direct-acting dopamine receptor drugs (agonist and antagonist). These results underscore the potential importance of nutritional status (e.g., glucose and insulin) in modulating dopamine neurotransmission and in so doing they begin to establish a neurochemical link between the high co-morbidity of eating disorders and drug abuse.
Collapse
Affiliation(s)
- Rajkumar J Sevak
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900, United States
| | | | | | | | | | | |
Collapse
|
47
|
Asada M, Ebihara S, Numachi Y, Okazaki T, Yamanda S, Ikeda K, Yasuda H, Sora I, Arai H. Reduced tumor growth in a mouse model of schizophrenia, lacking the dopamine transporter. Int J Cancer 2008; 123:511-8. [PMID: 18470912 DOI: 10.1002/ijc.23562] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The incidence of cancer in patients with schizophrenia has been reported to be lower that in the general population. On the other hand, it is well established that patients with schizophrenia have a hyper-dopaminergic system and dopamine has the ability to inhibit tumor angiogenesis. Therefore, in order to investigate the molecular mechanisms responsible for the lower cancer risk in schizophrenic patients, we used a mouse model of schizophrenia, which shows hyper-dopaminergic transmission in the nerve terminals of dopaminergic neurons. Here, we hypothesized that tumor growth was reduced in a mouse model of schizophrenia, lacking the dopamine transporter (DAT), and investigated tumor growth and angiogenesis in DAT knockout mice. The subcutaneous tumor in mice inoculated with cancer cells was smaller in DAT-/- mice than in the wild type (p < 0.05); however, the level of plasma dopamine in DAT-/- mice was lower than that of control littermates. Using human umbilical vascular endothelial cells (HUVEC), we examined dopamine signaling through dopamine D(1) receptor (D(1)R) and D(2)R. Dopamine stimulation slightly decreased the surface expression of vascular endothelial growth factor receptor-2 (VEGF-R2) but induced the phosphorylation of VEGF-R2 through Src in HUVEC. In addition, DAT-/- mice had less D(1)R. Both pharmacological and genetic interruption of D(1)R showed inhibited tumor growth. These results suggest that modulation of the dopaminergic system may contribute to cancer therapy.
Collapse
Affiliation(s)
- Masanori Asada
- Department of Geriatrics and Gerontology, Tohoku University School of Medicine, Sendai, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Thanos PK, Michaelides M, Benveniste H, Wang GJ, Volkow ND. The effects of cocaine on regional brain glucose metabolism is attenuated in dopamine transporter knockout mice. Synapse 2008; 62:319-24. [PMID: 18286542 DOI: 10.1002/syn.20503] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cocaine's ability to block the dopamine transporter (DAT) is crucial for its reinforcing effects. However the brain functional consequences of DAT blockade by cocaine are less clear since they are confounded by its concomitant blockade of norepinephrineand serotonin transporters. To separate the dopaminergic from the non-dopaminergic effects of cocaine on brain function we compared the regional brain metabolic responses to cocaine between dopamine transporter deficient (DAT(-/-)) mice with that of their DAT(+/+) littermates. We measured regional brain metabolism (marker of brain function) with 2-[18F]-fluoro-2-deoxy-D-glucose (FDG) and microPET (muPET) before and after acute cocaine administration (i.p. 10 mg/kg). Scans were conducted 2 weeks apart. At baseline DAT(-/-) mice had significantly greater metabolism in thalamus and cerebellum than DAT(+/+). Acute cocaine decreased whole brain metabolismand this effect was greater in DAT(+/+) (15%) than in DAT(-/-) mice (5%). DAT(+/+) mice showed regional decreases in the olfactory bulb, motor cortex, striatum, hippocampus, thalamus and cerebellum whereas DAT(-/-) mice showed decreases only in thalamus. The differential pattern of regional responses to cocaine in DAT(-/-) and DAT(+/+) suggests that most of the brain metabolic changes from acute cocaine are due to DAT blockade. Cocaine-induced decreases in metabolism in thalamus (region with dense noradrenergic innervation) in DAT(-/-) suggest that these were mediated by cocaine's blockade of norepinephrine transporters. The greater baseline metabolism in DAT(-/-) than DAT(+/+) mice in cerebellum (brain region mostly devoid of DAT) suggests that dopamine indirectly regulates activity of these brain regions.
Collapse
Affiliation(s)
- Panayotis K Thanos
- Behavioral Neuropharmacology Lab, Medical Department, Brookhaven National Laboratory, Upton, New York 11973-5000, USA.
| | | | | | | | | |
Collapse
|
49
|
Mattson BJ, Crombag HS, Mitchell T, Simmons DE, Kreuter JD, Morales M, Hope BT. Repeated amphetamine administration outside the home cage enhances drug-induced Fos expression in rat nucleus accumbens. Behav Brain Res 2007; 185:88-98. [PMID: 17720257 PMCID: PMC2135552 DOI: 10.1016/j.bbr.2007.07.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2007] [Revised: 07/09/2007] [Accepted: 07/15/2007] [Indexed: 12/28/2022]
Abstract
Induction of the immediate early gene protein product Fos has been used extensively to assess neural activation in the striatum after repeated amphetamine administration to rats in their home cages. However, this technique has not been used to examine striatal activation after repeated administration outside the home cage, an environment where repeated drug administration produces more robust psychomotor sensitization. We determined the dose-response relationship for amphetamine-induced psychomotor activity and Fos expression in nucleus accumbens and caudate-putamen 1 week after repeated administration of amphetamine or saline in locomotor activity chambers. Repeated administration of amphetamine enhanced amphetamine-induced locomotor activity and stereotypy and Fos expression in nucleus accumbens, but not in caudate-putamen. In comparison, levels of Fos expression induced by 1mg/kg amphetamine were not altered in nucleus accumbens or caudate-putamen by repeated amphetamine administration in the home cage. Double-labeling of Fos protein and enkephalin mRNA indicates that Fos is expressed in approximately equal numbers of enkephalin-negative and enkephalin-positive neurons in nucleus accumbens and caudate-putamen following injections outside the home cage. Furthermore, repeated amphetamine administration increased drug-induced Fos expression in enkephalin-positive, but not enkephalin-negative, neurons in nucleus accumbens. We conclude that repeated amphetamine administration outside the home cage recruits the activation of enkephalin-containing nucleus accumbens neurons during sensitized amphetamine-induced psychomotor activity.
Collapse
Affiliation(s)
- Brandi J Mattson
- Behavioral Neuroscience Branch, Intramural Research Program, The National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, 5500 Nathan Shock Drive, Baltimore, MD 21224, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Dopamine transporter mutant mice in experimental neuropharmacology. Naunyn Schmiedebergs Arch Pharmacol 2007; 377:301-13. [PMID: 18057916 DOI: 10.1007/s00210-007-0216-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2007] [Accepted: 11/02/2007] [Indexed: 12/27/2022]
Abstract
An opportunity to perform targeted genetic manipulations in mice has provided another dimension for modern pharmacological research. Genetically modified mice have become important tools to investigate functions of previously unexplored proteins, define mechanism of action of new and known pharmacological drugs, and validate novel targets for treatment of human disorders. One of the best examples of such use of genetic models in experimental pharmacology represents investigations involving mice deficient in the gene encoding the dopamine transporter (DAT). The dopamine transporter tightly regulates the extracellular dynamics of dopamine by recapturing released neurotransmitter into the presynaptic terminals, and genetic deletion of this protein results in profound alterations in both the presynaptic homeostasis and the extracellular dynamics of dopamine. By using this model of severe dopaminergic dysregulation, significant progress has been made in defining the major target of psychotropic drugs, understanding the mechanisms of their action, unraveling novel signaling events relevant for dopaminergic transmission, and mapping neuronal pathways involved in dopamine-related behaviors. Furthermore, DAT mutant mice provided an opportunity to model in vivo conditions of extreme dopaminergic dysfunction that could be relevant for human disorders such as ADHD, schizophrenia, and Parkinson's disease and, thus, could serve as test systems for developing novel treatments for these and related disorders.
Collapse
|