51
|
Rogers J, Renoir T, Hannan AJ. Gene-environment interactions informing therapeutic approaches to cognitive and affective disorders. Neuropharmacology 2017; 145:37-48. [PMID: 29277490 DOI: 10.1016/j.neuropharm.2017.12.038] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/17/2017] [Accepted: 12/20/2017] [Indexed: 02/06/2023]
Abstract
Gene-environment interactions drive experience-dependent changes in the brain that alter cognition, emotion and behaviour. Positive engagement with the environment, through novel experience and physical activity, can improve brain function, although the mechanisms mediating such experience-dependent plasticity remain to be fully elucidated. In this article, we discuss the therapeutic value of environmental stimuli, exercise and environmental enrichment (EE), for cognitive and affective disorders, with implications for the understanding and treatment of depression and anxiety disorders. We demonstrate that environmental manipulations are potential therapeutic strategies for improving outcomes in these psychiatric disorders, including beneficial impacts on cognition. We discuss how EE and exercise are therapeutic environmental interventions impacting both affective and cognitive function. Serotonergic (5-HTergic) signaling is strongly implicated in the manifestation of psychiatric disorders and regulates cognitive and emotional processing that can underpin them. Thus, we focus on evidence implicating the serotonergic system in mediating gene-environment interactions to EE and exercise. Finally, we discuss robust gene-environment interactions associated with EE and exercise interventions, and their impacts on specific brain areas, particularly the hippocampus. We focus on potential mediators of this experience-dependent plasticity, including adult neurogenesis and brain-derived neurotrophic factor (BDNF). Furthermore, we explore molecular and cellular mechanisms of experience-dependent plasticity that potentially underlie the restoration of affective and cognitive phenotypes, thus identifying novel therapeutic targets. This article is part of the Special Issue entitled "Neurobiology of Environmental Enrichment".
Collapse
Affiliation(s)
- Jake Rogers
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Australia
| | - Thibault Renoir
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Australia; Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Australia.
| |
Collapse
|
52
|
Hidalgo S, Molina-Mateo D, Escobedo P, Zárate RV, Fritz E, Fierro A, Perez EG, Iturriaga-Vasquez P, Reyes-Parada M, Varas R, Fuenzalida-Uribe N, Campusano JM. Characterization of a Novel Drosophila SERT Mutant: Insights on the Contribution of the Serotonin Neural System to Behaviors. ACS Chem Neurosci 2017; 8:2168-2179. [PMID: 28665105 DOI: 10.1021/acschemneuro.7b00089] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
A better comprehension on how different molecular components of the serotonergic system contribute to the adequate regulation of behaviors in animals is essential in the interpretation on how they are involved in neuropsychiatric and pathological disorders. It is possible to study these components in "simpler" animal models including the fly Drosophila melanogaster, given that most of the components of the serotonergic system are conserved between vertebrates and invertebrates. Here we decided to advance our understanding on how the serotonin plasma membrane transporter (SERT) contributes to serotonergic neurotransmission and behaviors in Drosophila. In doing this, we characterized for the first time a mutant for Drosophila SERT (dSERT) and additionally used a highly selective serotonin-releasing drug, 4-methylthioamphetamine (4-MTA), whose mechanism of action involves the SERT protein. Our results show that dSERT mutant animals exhibit an increased survival rate in stress conditions, increased basal motor behavior, and decreased levels in an anxiety-related parameter, centrophobism. We also show that 4-MTA increases the negative chemotaxis toward a strong aversive odorant, benzaldehyde. Our neurochemical data suggest that this effect is mediated by dSERT and depends on the 4-MTA-increased release of serotonin in the fly brain. Our in silico data support the idea that these effects are explained by specific interactions between 4-MTA and dSERT. In sum, our neurochemical, in silico, and behavioral analyses demonstrate the critical importance of the serotonergic system and particularly dSERT functioning in modulating several behaviors in Drosophila.
Collapse
Affiliation(s)
- Sergio Hidalgo
- Laboratorio
Neurogenética de la Conducta, Departamento de Biología
Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda #340, Santiago, Chile
| | - Daniela Molina-Mateo
- Laboratorio
Neurogenética de la Conducta, Departamento de Biología
Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda #340, Santiago, Chile
| | - Pía Escobedo
- Laboratorio
Neurogenética de la Conducta, Departamento de Biología
Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda #340, Santiago, Chile
| | - Rafaella V. Zárate
- Laboratorio
Neurogenética de la Conducta, Departamento de Biología
Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda #340, Santiago, Chile
| | - Elsa Fritz
- Laboratorio
Neurogenética de la Conducta, Departamento de Biología
Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda #340, Santiago, Chile
| | - Angélica Fierro
- Facultad
de Química, Pontificia Universidad Católica de Chile, Alameda #340, Santiago, Chile
| | - Edwin G. Perez
- Facultad
de Química, Pontificia Universidad Católica de Chile, Alameda #340, Santiago, Chile
| | | | - Miguel Reyes-Parada
- Escuela
de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Santiago, Chile
- Facultad
de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Rodrigo Varas
- Facultad
de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Nicolás Fuenzalida-Uribe
- Laboratorio
Neurogenética de la Conducta, Departamento de Biología
Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda #340, Santiago, Chile
| | - Jorge M. Campusano
- Laboratorio
Neurogenética de la Conducta, Departamento de Biología
Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda #340, Santiago, Chile
| |
Collapse
|
53
|
Fischer AG, Ullsperger M. An Update on the Role of Serotonin and its Interplay with Dopamine for Reward. Front Hum Neurosci 2017; 11:484. [PMID: 29075184 PMCID: PMC5641585 DOI: 10.3389/fnhum.2017.00484] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 09/19/2017] [Indexed: 01/02/2023] Open
Abstract
The specific role of serotonin and its interplay with dopamine (DA) in adaptive, reward guided behavior as well as drug dependance, still remains elusive. Recently, novel methods allowed cell type specific anatomical, functional and interventional analyses of serotonergic and dopaminergic circuits, promising significant advancement in understanding their functional roles. Furthermore, it is increasingly recognized that co-release of neurotransmitters is functionally relevant, understanding of which is required in order to interpret results of pharmacological studies and their relationship to neural recordings. Here, we review recent animal studies employing such techniques with the aim to connect their results to effects observed in human pharmacological studies and subjective effects of drugs. It appears that the additive effect of serotonin and DA conveys significant reward related information and is subjectively highly euphorizing. Neither DA nor serotonin alone have such an effect. This coincides with optogenetically targeted recordings in mice, where the dopaminergic system codes reward prediction errors (PE), and the serotonergic system mainly unsigned PE. Overall, this pattern of results indicates that joint activity between both systems carries essential reward information and invites parallel investigation of both neurotransmitter systems.
Collapse
Affiliation(s)
- Adrian G Fischer
- Department of Neuropsychology, Institute of Psychology, Otto-von-Guericke University, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Markus Ullsperger
- Department of Neuropsychology, Institute of Psychology, Otto-von-Guericke University, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany
| |
Collapse
|
54
|
Abdalla A, Atcherley CW, Pathirathna P, Samaranayake S, Qiang B, Peña E, Morgan SL, Heien ML, Hashemi P. In Vivo Ambient Serotonin Measurements at Carbon-Fiber Microelectrodes. Anal Chem 2017; 89:9703-9711. [PMID: 28795565 DOI: 10.1021/acs.analchem.7b01257] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The mechanisms that control extracellular serotonin levels in vivo are not well-defined. This shortcoming makes it very challenging to diagnose and treat the many psychiatric disorders in which serotonin is implicated. Fast-scan cyclic voltammetry (FSCV) can measure rapid serotonin release and reuptake events but cannot report critically important ambient serotonin levels. In this Article, we use fast-scan controlled adsorption voltammetry (FSCAV), to measure serotonin's steady-state, extracellular chemistry. We characterize the "Jackson" voltammetric waveform for FSCAV and show highly stable, selective, and sensitive ambient serotonin measurements in vitro. In vivo, we report basal serotonin levels in the CA2 region of the hippocampus as 64.9 ± 2.3 nM (n = 15 mice, weighted average ± standard error). We electrochemically and pharmacologically verify the selectivity of the serotonin signal. Finally, we develop a statistical model that incorporates the uncertainty in in vivo measurements, in addition to electrode variability, to more critically analyze the time course of pharmacological data. Our novel method is a uniquely powerful analysis tool that can provide deeper insights into the mechanisms that control serotonin's extracellular levels.
Collapse
Affiliation(s)
- Aya Abdalla
- Department of Chemistry and Biochemistry, University of South Carolina , 631 Sumter Street, Columbia, South Carolina 29208, United States
| | | | - Pavithra Pathirathna
- Department of Chemistry and Biochemistry, University of South Carolina , 631 Sumter Street, Columbia, South Carolina 29208, United States
| | - Srimal Samaranayake
- Department of Chemistry and Biochemistry, University of South Carolina , 631 Sumter Street, Columbia, South Carolina 29208, United States
| | - Beidi Qiang
- Department of Statistics, University of South Carolina , 1523 Greene Street, Columbia, South Carolina 29208, United States
| | - Edsel Peña
- Department of Statistics, University of South Carolina , 1523 Greene Street, Columbia, South Carolina 29208, United States
| | - Stephen L Morgan
- Department of Chemistry and Biochemistry, University of South Carolina , 631 Sumter Street, Columbia, South Carolina 29208, United States
| | - Michael L Heien
- Department of Chemistry and Biochemistry, University of Arizona , 1306 East University Blvd., Tucson, Arizona 85721, United States
| | - Parastoo Hashemi
- Department of Chemistry and Biochemistry, University of South Carolina , 631 Sumter Street, Columbia, South Carolina 29208, United States
| |
Collapse
|
55
|
PINHEIRO IL, SANTANA BJRCDE, GALINDO LCM, MANHÃES DE CASTRO R, SOUSA SLD. Perinatal serotonergic activity: A decisive factor in the control of food intake. REV NUTR 2017. [DOI: 10.1590/1678-98652017000400012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
ABSTRACT The serotoninergic system controls key events related to proper nervous system development. The neurotransmitter serotonin and the serotonin transporter are critical for this control. Availability of these components is minutely regulated during the development period, and the environment may affect their action on the nervous system. Environmental factors such as undernutrition and selective serotonin reuptake inhibitors may increase the availability of serotonin in the synaptic cleft and change its anorectic action. The physiological responses promoted by serotonin on intake control decrease when requested by acute stimuli or stress, demonstrating that animals or individuals develop adaptations in response to the environmental insults they experience during the development period. Diseases, such as anxiety and obesity, appear to be associated with the body’s response to stress or stimulus, and require greater serotonergic system action. These findings demonstrate the importance of the level of serotonin in the perinatal period to the development of molecular and morphological aspects of food intake control, and its decisive role in understanding the possible environmental factors that cause diseases in adulthood.
Collapse
|
56
|
Houwing DJ, Buwalda B, van der Zee EA, de Boer SF, Olivier JDA. The Serotonin Transporter and Early Life Stress: Translational Perspectives. Front Cell Neurosci 2017; 11:117. [PMID: 28491024 PMCID: PMC5405142 DOI: 10.3389/fncel.2017.00117] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 04/07/2017] [Indexed: 01/04/2023] Open
Abstract
The interaction between the serotonin transporter (SERT) linked polymorphic region (5-HTTLPR) and adverse early life stressing (ELS) events is associated with enhanced stress susceptibility and risk to develop mental disorders like major depression, anxiety, and aggressiveness. In particular, human short allele carriers are at increased risk. This 5-HTTLPR polymorphism is absent in the rodent SERT gene, but heterozygous SERT knockout rodents (SERT+/−) show several similarities to the human S-allele carrier, therefore creating an animal model of the human situation. Many rodent studies investigated ELS interactions in SERT knockout rodents combined with ELS. However, underlying neuromolecular mechanisms of the (mal)adaptive responses to adversity displayed by SERT rodents remain to be elucidated. Here, we provide a comprehensive review including studies describing mechanisms underlying SERT variation × ELS interactions in rodents. Alterations at the level of translation and transcription but also epigenetic alterations considerably contribute to underlying mechanisms of SERT variation × ELS interactions. In particular, SERT+/− rodents exposed to adverse early rearing environment may be of high translational and predictive value to the more stress sensitive human short-allele carrier, considering the similarity in neurochemical alterations. Therefore, SERT+/− rodents are highly relevant in research that aims to unravel the complex psychopathology of mental disorders. So far, most studies fail to show solid evidence for increased vulnerability to develop affective-like behavior after ELS in SERT+/− rodents. Several reasons may underlie these failures, e.g., (1) stressors used might not be optimal or severe enough to induce maladaptations, (2) effects in females are not sufficiently studied, and (3) few studies include both behavioral manifestations and molecular correlates of ELS-induced effects in SERT+/− rodents. Of course, one should not exclude the (although unlikely) possibility of SERT+/− rodents not being sensitive to ELS. In conclusion, future studies addressing ELS-induced effects in the SERT+/− rodents should extensively study both long-term behavioral and (epi)genetic aspects in both sexes. Finally, further research is warranted using more severe stressors in animal models. From there on, we should be able to draw solid conclusions whether the SERT+/− exposed to ELS is a suitable translational animal model for studying 5-HTTLPR polymorphism and stress interactions.
Collapse
Affiliation(s)
- Danielle J Houwing
- Unit Behavioral Neuroscience, Department of Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of GroningenGroningen, Netherlands
| | - Bauke Buwalda
- Unit Behavioral Neuroscience, Department of Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of GroningenGroningen, Netherlands
| | - Eddy A van der Zee
- Unit Molecular Neurobiology, Department of Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of GroningenGroningen, Netherlands
| | - Sietse F de Boer
- Unit Behavioral Neuroscience, Department of Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of GroningenGroningen, Netherlands
| | - Jocelien D A Olivier
- Unit Behavioral Neuroscience, Department of Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of GroningenGroningen, Netherlands
| |
Collapse
|
57
|
Zha W, Ho HTB, Hu T, Hebert MF, Wang J. Serotonin transporter deficiency drives estrogen-dependent obesity and glucose intolerance. Sci Rep 2017; 7:1137. [PMID: 28442777 PMCID: PMC5430688 DOI: 10.1038/s41598-017-01291-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 03/27/2017] [Indexed: 12/31/2022] Open
Abstract
Depression and use of antidepressant medications are both associated with increased risk of obesity, potentially attributed to a reduced serotonin transporter (SERT) function. However, how SERT deficiency promotes obesity is unknown. Here, we demonstrated that SERT−/− mice display abnormal fat accumulation in both white and brown adipose tissues, glucose intolerance and insulin resistance while exhibiting suppressed aromatase (Cyp19a1) expression and reduced circulating 17β-estradiol levels. 17β-estradiol replacement in SERT−/− mice reversed the obesity and glucose intolerance, supporting a role for estrogen in SERT deficiency-associated obesity and glucose intolerance. Treatment of wild type mice with paroxetine, a chemical inhibitor of SERT, also resulted in Cyp19a1 suppression, decreased circulating 17β-estradiol levels, abnormal fat accumulation, and glucose intolerance. Such effects were not observed in paroxetine-treated SERT−/− mice. Conversely, pregnant SERT−/− mice displayed normalized estrogen levels, markedly reduced fat accumulation, and improved glucose tolerance, which can be eliminated by an antagonist of estrogen receptor α (ERα). Together, these findings support that estrogen suppression is involved in SERT deficiency-induced obesity and glucose intolerance, and suggest approaches to restore 17β-estradiol levels as a novel treatment option for SERT deficiency associated obesity and metabolic abnormalities.
Collapse
Affiliation(s)
- Weibin Zha
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Horace T B Ho
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Tao Hu
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Mary F Hebert
- Department of Pharmacy, University of Washington, Seattle, WA, USA.,Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, USA
| | - Joanne Wang
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA. .,Nutrition Obesity Research Center, University of Washington, Seattle, WA, USA.
| |
Collapse
|
58
|
Neurodevelopmental Effects of Serotonin on the Brainstem Respiratory Network. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1015:193-216. [DOI: 10.1007/978-3-319-62817-2_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
59
|
van der Doelen RHA, Robroch B, Arnoldussen IA, Schulpen M, Homberg JR, Kozicz T. Serotonin and urocortin 1 in the dorsal raphe and Edinger-Westphal nuclei after early life stress in serotonin transporter knockout rats. Neuroscience 2016; 340:345-358. [PMID: 27826101 DOI: 10.1016/j.neuroscience.2016.10.072] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 10/29/2016] [Accepted: 10/31/2016] [Indexed: 02/04/2023]
Abstract
The interaction of early life stress (ELS) and the serotonin transporter (5-HTT) gene-linked polymorphic region (5-HTTLPR) has been associated with increased risk to develop depression in later life. We have used the maternal separation paradigm as a model for ELS exposure in homozygous and heterozygous 5-HTT knockout rats and measured urocortin 1 (Ucn1) mRNA and/or protein levels, Ucn1 DNA methylation, as well as 5-HT innervation in the centrally projecting Edinger-Westphal (EWcp) and dorsal raphe (DR) nuclei, both implicated in the regulation of stress response. We found that ELS and 5-HTT genotype increased the number of 5-HT neurons in specific DR subdivisions, and that 5-HTT knockout rats showed decreased 5-HT innervation of EWcp-Ucn1 neurons. Furthermore, ELS was associated with increased DNA methylation of the promoter region of the Ucn1 gene and increased expression of 5-HT receptor 1A in the EWcp. In contrast, 5-HTT deficiency was associated with site-specific alterations in DNA methylation of the Ucn1 promoter, and heterozygous 5-HTT knockout rats showed decreased expression of CRF receptor 1 in the EWcp. Together, our findings extend the existing literature on the relationship between EWcp-Ucn1 and DR-5-HT neurons. These observations will further our understanding on their potential contribution to mediate affect as a function of ELS interacting with 5-HTTLPR.
Collapse
Affiliation(s)
- Rick H A van der Doelen
- Department of Anatomy, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Berit Robroch
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ilse A Arnoldussen
- Department of Anatomy, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maya Schulpen
- Department of Anatomy, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Judith R Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tamás Kozicz
- Department of Anatomy, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Pediatrics, Hayward Genetics Center, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
60
|
Glover ME, Clinton SM. Of rodents and humans: A comparative review of the neurobehavioral effects of early life SSRI exposure in preclinical and clinical research. Int J Dev Neurosci 2016; 51:50-72. [PMID: 27165448 PMCID: PMC4930157 DOI: 10.1016/j.ijdevneu.2016.04.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 04/20/2016] [Accepted: 04/20/2016] [Indexed: 02/08/2023] Open
Abstract
Selective serotonin reuptake inhibitors (SSRIs) have been a mainstay pharmacological treatment for women experiencing depression during pregnancy and postpartum for the past 25 years. SSRIs act via blockade of the presynaptic serotonin transporter and result in a transient increase in synaptic serotonin. Long-lasting changes in cellular function such as serotonergic transmission, neurogenesis, and epigenetics, are thought to underlie the therapeutic benefits of SSRIs. In recent years, though, growing evidence in clinical and preclinical settings indicate that offspring exposed to SSRIs in utero or as neonates exhibit long-lasting behavioral adaptions. Clinically, children exposed to SSRIs in early life exhibit increased internalizing behavior reduced social behavior, and increased risk for depression in adolescence. Similarly, rodents exposed to SSRIs perinatally exhibit increased traits of anxiety- or depression-like behavior. Furthermore, certain individuals appear to be more susceptible to early life SSRI exposure than others, suggesting that perinatal SSRI exposure may pose greater risks for negative outcome within certain populations. Although SSRIs trigger a number of intracellular processes that likely contribute to their therapeutic effects, early life antidepressant exposure during critical neurodevelopmental periods may elicit lasting negative effects in offspring. In this review, we cover the basic development and structure of the serotonin system, how the system is affected by early life SSRI exposure, and the behavioral outcomes of perinatal SSRI exposure in both clinical and preclinical settings. We review recent evidence indicating that perinatal SSRI exposure perturbs the developing limbic system, including altered serotonergic transmission, neurogenesis, and epigenetic processes in the hippocampus, which may contribute to behavioral domains (e.g., sociability, cognition, anxiety, and behavioral despair) that are affected by perinatal SSRI treatment. Identifying the molecular mechanisms that underlie the deleterious behavioral effects of perinatal SSRI exposure may highlight biological mechanisms in the etiology of mood disorders. Moreover, because recent studies suggest that certain individuals may be more susceptible to the negative consequences of early life SSRI exposure than others, understanding mechanisms that drive such susceptibility could lead to individualized treatment strategies for depressed women who are or plan to become pregnant.
Collapse
Affiliation(s)
| | - Sarah M Clinton
- Department of Psychiatry, University of Alabama-Birmingham, USA.
| |
Collapse
|
61
|
Koopman KE, Roefs A, Elbers DCE, Fliers E, Booij J, Serlie MJ, la Fleur SE. Brain dopamine and serotonin transporter binding are associated with visual attention bias for food in lean men. Psychol Med 2016; 46:1707-1717. [PMID: 26984412 DOI: 10.1017/s0033291716000222] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND In rodents, the striatal dopamine (DA) system and the (hypo)thalamic serotonin (5-HT) system are involved in the regulation of feeding behavior. In lean humans, little is known about the relationship between these brain neurotransmitter systems and feeding. We studied the relationship between striatal DA transporters (DAT) and diencephalic 5-HT transporters (SERT), behavioral tasks and questionnaires, and food intake. METHOD We measured striatal DAT and diencephalic SERT binding with [123I]FP-CIT SPECT in 36 lean male subjects. Visual attention bias for food (detection speed and distraction time) and degree of impulsivity were measured using response-latency-based computer tasks. Craving and emotional eating were assessed with questionnaires and ratings of hunger by means of VAS scores. Food intake was assessed through a self-reported online diet journal. RESULTS Striatal DAT and diencephalic SERT binding negatively correlated with food detection speed (p = 0.008, r = -0.50 and p = 0.002, r = -0.57, respectively), but not with food distraction time, ratings of hunger, craving or impulsivity. Striatal DAT and diencephalic SERT binding did not correlate with free choice food intake, whereas food detection speed positively correlated with total caloric intake (p = 0.001, r = 0.60), protein intake (p = 0.01, r = 0.44), carbohydrate intake (p = 0.03, r = 0.39) and fat intake (p = 0.06, r = 0.35). CONCLUSIONS These results indicate a role for the central 5-HT and DA system in the regulation of visual attention bias for food, which contributes to the motivation to eat, in non-obese, healthy humans. In addition, this study confirms that food detection speed, measured with the latency-based computer task, positively correlates with total food and macronutrient intake.
Collapse
Affiliation(s)
- K E Koopman
- Department of Endocrinology & Metabolism,Academic Medical Center Amsterdam,University of Amsterdam,The Netherlands
| | - A Roefs
- Faculty of Psychology & Neuroscience,Maastricht University,Maastricht,The Netherlands
| | - D C E Elbers
- Department of Endocrinology & Metabolism,Academic Medical Center Amsterdam,University of Amsterdam,The Netherlands
| | - E Fliers
- Department of Endocrinology & Metabolism,Academic Medical Center Amsterdam,University of Amsterdam,The Netherlands
| | - J Booij
- Department of Nuclear Medicine,Academic Medical Center Amsterdam,University of Amsterdam,The Netherlands
| | - M J Serlie
- Department of Endocrinology & Metabolism,Academic Medical Center Amsterdam,University of Amsterdam,The Netherlands
| | - S E la Fleur
- Department of Endocrinology & Metabolism,Academic Medical Center Amsterdam,University of Amsterdam,The Netherlands
| |
Collapse
|
62
|
Meyer N, Richter SH, Schreiber RS, Kloke V, Kaiser S, Lesch KP, Sachser N. The Unexpected Effects of Beneficial and Adverse Social Experiences during Adolescence on Anxiety and Aggression and Their Modulation by Genotype. Front Behav Neurosci 2016; 10:97. [PMID: 27303275 PMCID: PMC4880570 DOI: 10.3389/fnbeh.2016.00097] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 05/06/2016] [Indexed: 12/28/2022] Open
Abstract
Anxiety and aggression are part of the behavioral repertoire of humans and animals. However, in their exaggerated form both can become maladaptive and result in psychiatric disorders. On the one hand, genetic predisposition has been shown to play a crucial modulatory role in anxiety and aggression. On the other hand, social experiences have been implicated in the modulation of these traits. However, so far, mainly experiences in early life phases have been considered crucial for shaping anxiety-like and aggressive behavior, while the phase of adolescence has largely been neglected. Therefore, the aim of the present study was to elucidate how levels of anxiety-like and aggressive behavior are shaped by social experiences during adolescence and serotonin transporter (5-HTT) genotype. For this purpose, male mice of a 5-HTT knockout mouse model including all three genotypes (wildtype, heterozygous and homozygous 5-HTT knockout mice) were either exposed to an adverse social situation or a beneficial social environment during adolescence. This was accomplished in a custom-made cage system where mice experiencing the adverse environment were repeatedly introduced to the territory of a dominant opponent but had the possibility to escape to a refuge cage. Mice encountering beneficial social conditions had free access to a female mating partner. Afterwards, anxiety-like and aggressive behavior was assessed in a battery of tests. Surprisingly, unfavorable conditions during adolescence led to a decrease in anxiety-like behavior and an increase in exploratory locomotion. Additionally, aggressive behavior was augmented in animals that experienced social adversity. Concerning genotype, homozygous 5-HTT knockout mice were more anxious and less aggressive than heterozygous 5-HTT knockout and wildtype mice. In summary, adolescence is clearly an important phase in which anxiety-like and aggressive behavior can be shaped. Furthermore, it seems that having to cope with challenge during adolescence instead of experiencing throughout beneficial social conditions leads to reduced levels of anxiety-like behavior.
Collapse
Affiliation(s)
- Neele Meyer
- Department of Behavioural Biology, University of MuensterMuenster, Germany; Muenster Graduate School of Evolution, University of MuensterMuenster, Germany
| | - S Helene Richter
- Department of Behavioural Biology, University of Muenster Muenster, Germany
| | | | - Vanessa Kloke
- Department of Behavioural Biology, University of Muenster Muenster, Germany
| | - Sylvia Kaiser
- Department of Behavioural Biology, University of MuensterMuenster, Germany; Muenster Graduate School of Evolution, University of MuensterMuenster, Germany
| | - Klaus-Peter Lesch
- Laboratory of Translational Neuroscience, Division of Molecular Psychiatry, Department of Psychiatry, Psychosomatics, and Psychotherapy, University of Wuerzburg Wuerzburg, Germany
| | - Norbert Sachser
- Department of Behavioural Biology, University of MuensterMuenster, Germany; Muenster Graduate School of Evolution, University of MuensterMuenster, Germany
| |
Collapse
|
63
|
Bocchio M, McHugh SB, Bannerman DM, Sharp T, Capogna M. Serotonin, Amygdala and Fear: Assembling the Puzzle. Front Neural Circuits 2016; 10:24. [PMID: 27092057 PMCID: PMC4820447 DOI: 10.3389/fncir.2016.00024] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 03/21/2016] [Indexed: 11/13/2022] Open
Abstract
The fear circuitry orchestrates defense mechanisms in response to environmental threats. This circuitry is evolutionarily crucial for survival, but its dysregulation is thought to play a major role in the pathophysiology of psychiatric conditions in humans. The amygdala is a key player in the processing of fear. This brain area is prominently modulated by the neurotransmitter serotonin (5-hydroxytryptamine, 5-HT). The 5-HT input to the amygdala has drawn particular interest because genetic and pharmacological alterations of the 5-HT transporter (5-HTT) affect amygdala activation in response to emotional stimuli. Nonetheless, the impact of 5-HT on fear processing remains poorly understood.The aim of this review is to elucidate the physiological role of 5-HT in fear learning via its action on the neuronal circuits of the amygdala. Since 5-HT release increases in the basolateral amygdala (BLA) during both fear memory acquisition and expression, we examine whether and how 5-HT neurons encode aversive stimuli and aversive cues. Next, we describe pharmacological and genetic alterations of 5-HT neurotransmission that, in both rodents and humans, lead to altered fear learning. To explore the mechanisms through which 5-HT could modulate conditioned fear, we focus on the rodent BLA. We propose that a circuit-based approach taking into account the localization of specific 5-HT receptors on neurochemically-defined neurons in the BLA may be essential to decipher the role of 5-HT in emotional behavior. In keeping with a 5-HT control of fear learning, we review electrophysiological data suggesting that 5-HT regulates synaptic plasticity, spike synchrony and theta oscillations in the BLA via actions on different subcellular compartments of principal neurons and distinct GABAergic interneuron populations. Finally, we discuss how recently developed optogenetic tools combined with electrophysiological recordings and behavior could progress the knowledge of the mechanisms underlying 5-HT modulation of fear learning via action on amygdala circuits. Such advancement could pave the way for a deeper understanding of 5-HT in emotional behavior in both health and disease.
Collapse
Affiliation(s)
- Marco Bocchio
- MRC Brain Network Dynamics Unit, Department of Pharmacology, University of Oxford Oxford, UK
| | - Stephen B McHugh
- Department of Experimental Psychology, University of Oxford Oxford, UK
| | - David M Bannerman
- Department of Experimental Psychology, University of Oxford Oxford, UK
| | - Trevor Sharp
- Department of Pharmacology, University of Oxford Oxford, UK
| | - Marco Capogna
- MRC Brain Network Dynamics Unit, Department of Pharmacology, University of Oxford Oxford, UK
| |
Collapse
|
64
|
Ye R, Quinlan MA, Iwamoto H, Wu HH, Green NH, Jetter CS, McMahon DG, Veestra-VanderWeele J, Levitt P, Blakely RD. Physical Interactions and Functional Relationships of Neuroligin 2 and Midbrain Serotonin Transporters. Front Synaptic Neurosci 2016; 7:20. [PMID: 26793096 PMCID: PMC4707279 DOI: 10.3389/fnsyn.2015.00020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 12/14/2015] [Indexed: 12/31/2022] Open
Abstract
The neurotransmitter serotonin [5-hydroxytryptamine (5-HT)] modulates many key brain functions including those subserving sensation, emotion, reward, and cognition. Efficient clearance of 5-HT after release is achieved by the antidepressant-sensitive 5-HT transporter (SERT, SLC6A4). To identify novel SERT regulators, we pursued a proteomic analysis of mouse midbrain SERT complexes, evaluating findings in the context of prior studies that established a SERT-linked transcriptome. Remarkably, both efforts converged on a relationship of SERT with the synaptic adhesion protein neuroligin 2 (NLGN2), a post-synaptic partner for presynaptic neurexins, and a protein well-known to organize inhibitory GABAergic synapses. Western blots of midbrain reciprocal immunoprecipitations confirmed SERT/NLGN2 associations, and also extended to other NLGN2 associated proteins [e.g., α-neurexin (NRXN), gephyrin]. Midbrain SERT/NLGN2 interactions were found to be Ca(2+)-independent, supporting cis vs. trans-synaptic interactions, and were absent in hippocampal preparations, consistent with interactions arising in somatodendritic compartments. Dual color in situ hybridization confirmed co-expression of Tph2 and Nlgn2 mRNA in the dorsal raphe, with immunocytochemical studies confirming SERT:NLGN2 co-localization in raphe cell bodies but not axons. Consistent with correlative mRNA expression studies, loss of NLGN2 expression in Nlgn2 null mice produced significant reductions in midbrain and hippocampal SERT expression and function. Additionally, dorsal raphe 5-HT neurons from Nlgn2 null mice exhibit reduced excitability, a loss of GABAA receptor-mediated IPSCs, and increased 5-HT1A autoreceptor sensitivity. Finally, Nlgn2 null mice display significant changes in behaviors known to be responsive to SERT and/or 5-HT receptor manipulations. We discuss our findings in relation to the possible coordination of intrinsic and extrinsic regulation afforded by somatodendritic SERT:NLGN2 complexes.
Collapse
Affiliation(s)
- Ran Ye
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville TN, USA
| | - Meagan A Quinlan
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville TN, USA
| | - Hideki Iwamoto
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville TN, USA
| | - Hsiao-Huei Wu
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville TN, USA
| | - Noah H Green
- Department of Biological Sciences, Vanderbilt University School of Medicine, Nashville TN, USA
| | - Christopher S Jetter
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville TN, USA
| | - Douglas G McMahon
- Department of Pharmacology, Vanderbilt University School of Medicine, NashvilleTN, USA; Department of Biological Sciences, Vanderbilt University School of Medicine, NashvilleTN, USA
| | - Jeremy Veestra-VanderWeele
- Department of Psychiatry, NYS Psychiatric Institute, Columbia University Medical Center, New York NY, USA
| | - Pat Levitt
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville TN, USA
| | - Randy D Blakely
- Department of Pharmacology, Vanderbilt University School of Medicine, NashvilleTN, USA; Department of Psychiatry, Vanderbilt University School of Medicine, NashvilleTN, USA
| |
Collapse
|
65
|
Hvoslef-Eide M, Nilsson SRO, Saksida LM, Bussey TJ. Cognitive Translation Using the Rodent Touchscreen Testing Approach. Curr Top Behav Neurosci 2016; 28:423-447. [PMID: 27305921 DOI: 10.1007/7854_2015_5007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The development of novel therapeutic avenues for the treatment of cognitive deficits in psychiatric and neurodegenerative disease is of high importance, yet progress in this field has been slow. One reason for this lack of success may lie in discrepancies between how cognitive functions are assessed in experimental animals and humans. In an attempt to bridge this translational gap, the rodent touchscreen testing platform is suggested as a translational tool. Specific examples of successful cross-species translation are discussed focusing on paired associate learning (PAL), the 5-choice serial reaction time task (5-CSRTT), the rodent continuous performance task (rCPT) and reversal learning. With ongoing research assessing the neurocognitive validity of tasks, the touchscreen approach is likely to become increasingly prevalent in translational cognitive research.
Collapse
Affiliation(s)
- M Hvoslef-Eide
- Department of Psychology, University of Cambridge, Cambridge, CB2 3EB, UK.
| | - S R O Nilsson
- Department of Psychology, University of Cambridge, Cambridge, CB2 3EB, UK
| | - L M Saksida
- Department of Psychology, University of Cambridge, Cambridge, CB2 3EB, UK
| | - T J Bussey
- Department of Psychology, University of Cambridge, Cambridge, CB2 3EB, UK
| |
Collapse
|
66
|
Harrison SJ, Tyrer AE, Levitan RD, Xu X, Houle S, Wilson AA, Nobrega JN, Rusjan PM, Meyer JH. Light therapy and serotonin transporter binding in the anterior cingulate and prefrontal cortex. Acta Psychiatr Scand 2015; 132:379-88. [PMID: 25891484 PMCID: PMC4942271 DOI: 10.1111/acps.12424] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/17/2015] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To investigate the effects of light therapy on serotonin transporter binding (5-HTT BPND ), an index of 5-HTT levels, in the anterior cingulate and prefrontal cortices (ACC and PFC) of healthy individuals during the fall and winter. Twenty-five per cent of healthy individuals experience seasonal mood changes that affect functioning. 5-HTT BPND has been found to be higher across multiple brain regions in the fall and winter relative to spring and summer, and elevated 5-HTT BPND may lead to extracellular serotonin loss and low mood. We hypothesized that, during the fall and winter, light therapy would reduce 5-HTT BPND in the ACC and PFC, which sample brain regions involved in mood regulation. METHOD In a single-blind, placebo-controlled, counterbalanced, crossover design, [(11) C]DASB positron emission tomography was used measure 5-HTT BPND following light therapy and placebo conditions during fall and winter. RESULTS In winter, light therapy significantly decreased 5-HTT BPND by 12% in the ACC relative to placebo (F1,9 = 18.04, P = 0.002). In the fall, no significant change in 5-HTT BPND was found in any region across conditions. CONCLUSION These results identify, for the first time, a central biomarker associated with the intervention of light therapy in humans which may be applied to further develop this treatment for prevention of seasonal depression.
Collapse
Affiliation(s)
- S J Harrison
- CAMH Research Imaging Centre and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health and Departments of Psychiatry, Pharmacology and Toxicology, and Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Behavioural Neurobiology Laboratory and Campbell Family Mental Health Research Institute and Departments of Psychiatry, Pharmacology and Toxicology, and Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - A E Tyrer
- CAMH Research Imaging Centre and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health and Departments of Psychiatry, Pharmacology and Toxicology, and Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - R D Levitan
- CAMH Research Imaging Centre and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health and Departments of Psychiatry, Pharmacology and Toxicology, and Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - X Xu
- CAMH Research Imaging Centre and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health and Departments of Psychiatry, Pharmacology and Toxicology, and Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - S Houle
- CAMH Research Imaging Centre and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health and Departments of Psychiatry, Pharmacology and Toxicology, and Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - A A Wilson
- CAMH Research Imaging Centre and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health and Departments of Psychiatry, Pharmacology and Toxicology, and Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - J N Nobrega
- CAMH Research Imaging Centre and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health and Departments of Psychiatry, Pharmacology and Toxicology, and Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Behavioural Neurobiology Laboratory and Campbell Family Mental Health Research Institute and Departments of Psychiatry, Pharmacology and Toxicology, and Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - P M Rusjan
- CAMH Research Imaging Centre and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health and Departments of Psychiatry, Pharmacology and Toxicology, and Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - J H Meyer
- CAMH Research Imaging Centre and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health and Departments of Psychiatry, Pharmacology and Toxicology, and Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
67
|
Yuan ZX, Rapoport SI. Transient postnatal fluoxetine decreases brain concentrations of 20-HETE and 15-epi-LXA4, arachidonic acid metabolites in adult mice. Prostaglandins Leukot Essent Fatty Acids 2015; 101:9-14. [PMID: 26234927 PMCID: PMC4581970 DOI: 10.1016/j.plefa.2015.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 07/11/2015] [Accepted: 07/13/2015] [Indexed: 01/17/2023]
Abstract
BACKGROUND Transient postnatal exposure of rodents to the selective serotonin (5-HT) reuptake inhibitor (SSRI) fluoxetine alters behavior and brain 5-HT neurotransmission during adulthood, and also reduces brain arachidonic (ARA) metabolic consumption and protein level of the ARA metabolizing enzyme, cytochrome P4504A (CYP4A). HYPOTHESIS Brain 20-hydroxyeicosatetraenoic acid (20-HETE), converted by CYP4A from ARA, will be reduced in adult mice treated transiently and postnatally with fluoxetine. METHODS Male mice pups were injected i.p. daily with fluoxetine (10mg/kg) or saline during P4-P21. At P90 their brain was high-energy microwaved and analyzed for 20-HETE and six other ARA metabolites by enzyme immunoassay. RESULTS Postnatal fluoxetine vs. saline significantly decreased brain concentrations of 20-HETE (-70.3%) and 15-epi-lipoxin A4 (-60%) in adult mice, but did not change other eicosanoid concentrations. CONCLUSIONS Behavioral changes in adult mice treated postnatally with fluoxetine may be related to reduced brain ARA metabolism involving CYP4A and 20-HETE formation.
Collapse
Affiliation(s)
- Zhi-Xin Yuan
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Stanley I Rapoport
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
68
|
Attenuated methamphetamine-induced locomotor sensitization in serotonin transporter knockout mice is restored by serotonin 1B receptor antagonist treatment. Behav Pharmacol 2015; 26:167-79. [PMID: 25485646 DOI: 10.1097/fbp.0000000000000120] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Repeated administration of methamphetamine (METH) enhances acute locomotor responses to METH administered in the same context, a phenomenon termed as 'locomotor sensitization'. Although many of the acute effects of METH are mediated by its influences on the compartmentalization of dopamine, serotonin systems have also been suggested to influence the behavioral effects of METH in ways that are not fully understood. The present experiments examined serotonergic roles in METH-induced locomotor sensitization by assessing: (a) the effect of serotonin transporter (SERT; Slc6A4) knockout (KO) on METH-induced locomotor sensitization; (b) extracellular monoamine levels in METH-treated animals as determined by in-vivo microdialysis; and (c) effects of serotonin (5-HT) receptor antagonists on METH-induced behavioral sensitization, with focus on effects of the 5-HT1B receptor antagonist SB 216641 and a comparison with the 5-HT2 receptor antagonist ketanserin. Repeated METH administration failed to induce behavioral sensitization in homozygous SERT KO (SERT-/-) mice under conditions that produced substantial sensitization in wild-type or heterozygous SERT KO (SERT+/-) mice. The selective 5-HT1B antagonist receptor SB 216641 restored METH-induced locomotor sensitization in SERT-/- mice, whereas ketanserin was ineffective. METH-induced increases in extracellular 5-HT (5-HTex) levels were substantially reduced in SERT-/- mice, although SERT genotype had no effect on METH-induced increases in extracellular dopamine. These experiments demonstrate that 5-HT actions, including those at 5-HT1B receptors, contribute to METH-induced locomotor sensitization. Modulation of 5-HT1B receptors might aid therapeutic approaches to the sequelae of chronic METH use.
Collapse
|
69
|
Frazer S, Otomo K, Dayer A. Early-life serotonin dysregulation affects the migration and positioning of cortical interneuron subtypes. Transl Psychiatry 2015; 5:e644. [PMID: 26393490 PMCID: PMC5068808 DOI: 10.1038/tp.2015.147] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 07/22/2015] [Accepted: 08/11/2015] [Indexed: 12/21/2022] Open
Abstract
Early-life deficiency of the serotonin transporter (SERT) gives rise to a wide range of psychiatric-relevant phenotypes; however, the molecular and cellular targets of serotonin dyregulation during neural circuit formation remain to be identified. Interestingly, migrating cortical interneurons (INs) derived from the caudal ganglionic eminence (CGE) have been shown to be more responsive to serotonin-mediated signalling compared with INs derived from the medial ganglionic eminence (MGE). Here we investigated the impact of early-life SERT deficiency on the migration and positioning of CGE-derived cortical INs in SERT-ko mice and in mice exposed to the SERT inhibitor fluoxetine during the late embryonic period. Using confocal time-lapse imaging and microarray-based expression analysis we found that genetic and pharmacological SERT deficiency significantly increased the migratory speed of CGE-derived INs and affected transcriptional programmes regulating neuronal migration. Postnatal studies revealed that SERT deficiency altered the cortical laminar distribution of subtypes of CGE-derived INs but not MGE-derived INs. More specifically, we found that the distribution of vasointestinal peptide (VIP)-expressing INs in layer 2/3 was abnormal in both genetic and pharmacological SERT-deficiency models. Collectively, these data indicate that early-life SERT deficiency has an impact on the migration and molecular programmes of CGE-derived INs, thus leading to specific alterations in the positioning of VIP-expressing INs. These data add to the growing evidence that early-life serotonin dysregulation affects cortical microcircuit formation and contributes to the emergence of psychiatric-relevant phenotypes.
Collapse
Affiliation(s)
- S Frazer
- Department of Mental Health and Psychiatry, University of Geneva Medical School, Geneva, Switzerland,Department of Psychiatry and Basic Neurosciences, University of Geneva Medical School, Geneva, Switzerland
| | - K Otomo
- Department of Mental Health and Psychiatry, University of Geneva Medical School, Geneva, Switzerland,Department of Psychiatry and Basic Neurosciences, University of Geneva Medical School, Geneva, Switzerland
| | - A Dayer
- Department of Mental Health and Psychiatry, University of Geneva Medical School, Geneva, Switzerland,Department of Psychiatry and Basic Neurosciences, University of Geneva Medical School, Geneva, Switzerland,Department of Psychiatry and Basic Neurosciences, University of Geneva Medical School (CMU), Rue Michel-Servet 1, 1211 Genève 4, Geneva 1211, Switzerland. E-mail:
| |
Collapse
|
70
|
Serotonin reuptake inhibitors and serotonin transporter genotype modulate performance monitoring functions but not their electrophysiological correlates. J Neurosci 2015; 35:8181-90. [PMID: 26019334 DOI: 10.1523/jneurosci.5124-14.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Serotonin (5-HT) has been hypothesized to be implicated in performance monitoring by promoting behavioral inhibition in the face of aversive events. However, it is unclear whether this is restricted to external (punishment) or includes internal (response errors) events. The aim of the current study was to test whether higher 5-HT levels instigate inhibition specifically in the face of errors, measured as post-error slowing (PES), and whether this is represented in electrophysiological correlates of error processing, namely error-related negativity (ERN) and positivity. Therefore, from a large sample of human subjects (n = 878), two extreme groups were formed regarding hypothesized high and low 5-HT transporter (5-HTT) expression based on 5-HTTLPR and two additional single nucleotide polymorphisms (rs25531, rs25532). Seventeen higher (LL) and 15 lower (SS) expressing Caucasian subjects were administered the selective serotonin reuptake inhibitor (SSRI) citalopram (10 mg) intravenously in a double-blind crossover design. We found pharmacogenetic evidence for a role of 5-HT in mediating PES: SSRI administration increased PES in both genetic groups, and SS subjects displayed higher PES. These effects were absent on post-conflict slowing. However, ERN and error positivity were unaffected by pharmacogenetic factors, but ERN was decoupled from behavioral adaptation by SSRI administration in the LL group. Thus, pharmacogenetic evidence suggests that increased 5-HT levels lead to behavioral inhibition in the context of internal aversive events, but electrophysiological correlates of performance monitoring appear unrelated to the 5-HT system. Therefore, our findings are consistent with theories suggesting that 5-HT mediates the link between aversive processing and inhibition.
Collapse
|
71
|
Yang H, Sampson MM, Senturk D, Andrews AM. Sex- and SERT-mediated differences in stimulated serotonin revealed by fast microdialysis. ACS Chem Neurosci 2015; 6:1487-501. [PMID: 26167657 DOI: 10.1021/acschemneuro.5b00132] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In vivo microdialysis is widely used to investigate how neurotransmitter levels in the brain respond to biologically relevant challenges. Here, we combined recent improvements in the temporal resolution of online sampling and analysis for serotonin with a brief high-K(+) stimulus paradigm to study the dynamics of evoked release. We observed stimulated serotonin overflow with high-K(+) pulses as short as 1 min when determined with 2-min dialysate sampling in ventral striatum. Stimulated serotonin levels in female mice during the high estrogen period of the estrous cycle were similar to serotonin levels in male mice. By contrast, stimulated serotonin overflow during the low estrogen period in female mice was increased to levels similar to those in male mice with local serotonin transporter (SERT) inhibition. Stimulated serotonin levels in mice with constitutive loss of SERT were considerably higher yet, pointing to neuroadaptive potentiation of serotonin release. When combined with brief K(+) stimulation, fast microdialysis reveals dynamic changes in extracellular serotonin levels associated with normal hormonal cycles and pharmacologic vs genetic loss of SERT function.
Collapse
Affiliation(s)
- Hongyan Yang
- Department of Psychiatry & Biobehavioral Sciences, Hatos Center for Neuropharmacology, Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine, ‡Molecular Toxicology Interdepartmental Program, §Department of Chemistry & Biochemistry, and ∥Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Maureen M. Sampson
- Department of Psychiatry & Biobehavioral Sciences, Hatos Center for Neuropharmacology, Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine, ‡Molecular Toxicology Interdepartmental Program, §Department of Chemistry & Biochemistry, and ∥Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Damla Senturk
- Department of Psychiatry & Biobehavioral Sciences, Hatos Center for Neuropharmacology, Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine, ‡Molecular Toxicology Interdepartmental Program, §Department of Chemistry & Biochemistry, and ∥Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Anne M. Andrews
- Department of Psychiatry & Biobehavioral Sciences, Hatos Center for Neuropharmacology, Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine, ‡Molecular Toxicology Interdepartmental Program, §Department of Chemistry & Biochemistry, and ∥Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California 90095, United States
| |
Collapse
|
72
|
Schipper P, Lopresto D, Reintjes RJ, Joosten J, Henckens MJAG, Kozicz T, Homberg JR. Improved Stress Control in Serotonin Transporter Knockout Rats: Involvement of the Prefrontal Cortex and Dorsal Raphe Nucleus. ACS Chem Neurosci 2015; 6:1143-50. [PMID: 26132384 DOI: 10.1021/acschemneuro.5b00126] [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] [Indexed: 11/29/2022] Open
Abstract
Variations in serotonin transporter (5-HTT) expression have been associated with altered sensitivity to stress. Since controllability is known to alter the impact of a stressor through differential activation of the medial prefrontal cortex (mPFC) and dorsal raphe nucleus (DRN), and that these regions are functionally affected by genetic 5-HTT down-regulation, we hypothesized that 5-HTT expression modulates the effect of controllability on stressor impact and coping. Here, we investigated the effects of a signaled stress controllability task or a yoked uncontrollable stressor on behavioral responding and mPFC and DRN activation. 5-HTT(-/-) rats proved better capable of acquiring the active avoidance task than 5-HTT(+/+) animals. Controllability determined DRN activation in 5-HTT(+/+), but not 5-HTT(-/-), rats, whereas controllability-related activation of the mPFC was independent of genotype. These findings suggest that serotonergic activation in the DRN is involved in stress coping in a 5-HTT expression dependent manner, whereas mPFC activation seems to be implicated in control over stress independently of 5-HTT expression. We speculate that alterations in serotonergic feedback in the DRN might be a potential mechanism driving this differential stress coping.
Collapse
Affiliation(s)
- Pieter Schipper
- Donders Institute for Brain, Cognition and Behaviour,
Centre for Neuroscience, Department of Cognitive Neuroscience, and ‡Donders
Institute for Brain, Cognition and Behaviour, Centre for Neuroscience,
Department of Anatomy, Radboud University Medical Centre, Geert
Grooteplein 21, 6525 EZ Nijmegen, The Netherlands
| | - Dora Lopresto
- Donders Institute for Brain, Cognition and Behaviour,
Centre for Neuroscience, Department of Cognitive Neuroscience, and ‡Donders
Institute for Brain, Cognition and Behaviour, Centre for Neuroscience,
Department of Anatomy, Radboud University Medical Centre, Geert
Grooteplein 21, 6525 EZ Nijmegen, The Netherlands
| | - Roy J. Reintjes
- Donders Institute for Brain, Cognition and Behaviour,
Centre for Neuroscience, Department of Cognitive Neuroscience, and ‡Donders
Institute for Brain, Cognition and Behaviour, Centre for Neuroscience,
Department of Anatomy, Radboud University Medical Centre, Geert
Grooteplein 21, 6525 EZ Nijmegen, The Netherlands
| | - Joep Joosten
- Donders Institute for Brain, Cognition and Behaviour,
Centre for Neuroscience, Department of Cognitive Neuroscience, and ‡Donders
Institute for Brain, Cognition and Behaviour, Centre for Neuroscience,
Department of Anatomy, Radboud University Medical Centre, Geert
Grooteplein 21, 6525 EZ Nijmegen, The Netherlands
| | - Marloes J. A. G. Henckens
- Donders Institute for Brain, Cognition and Behaviour,
Centre for Neuroscience, Department of Cognitive Neuroscience, and ‡Donders
Institute for Brain, Cognition and Behaviour, Centre for Neuroscience,
Department of Anatomy, Radboud University Medical Centre, Geert
Grooteplein 21, 6525 EZ Nijmegen, The Netherlands
| | - Tamas Kozicz
- Donders Institute for Brain, Cognition and Behaviour,
Centre for Neuroscience, Department of Cognitive Neuroscience, and ‡Donders
Institute for Brain, Cognition and Behaviour, Centre for Neuroscience,
Department of Anatomy, Radboud University Medical Centre, Geert
Grooteplein 21, 6525 EZ Nijmegen, The Netherlands
| | - Judith R. Homberg
- Donders Institute for Brain, Cognition and Behaviour,
Centre for Neuroscience, Department of Cognitive Neuroscience, and ‡Donders
Institute for Brain, Cognition and Behaviour, Centre for Neuroscience,
Department of Anatomy, Radboud University Medical Centre, Geert
Grooteplein 21, 6525 EZ Nijmegen, The Netherlands
| |
Collapse
|
73
|
Dayer AG, Jacobshagen M, Chaumont-Dubel S, Marin P. 5-HT6 Receptor: A New Player Controlling the Development of Neural Circuits. ACS Chem Neurosci 2015; 6:951-60. [PMID: 25590789 DOI: 10.1021/cn500326z] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
5-HT6 receptor (5-HT6R) is a G protein-coupled receptor that has recently emerged as a new regulator of neural development. In addition to the canonical Gs adenylyl cyclase pathway, recent proteomics approaches reveal that 5-HT6R is able to engage key developmental signaling pathways controlling neuronal circuit formation, neuronal connectivity, and psychiatric-relevant behaviors. For example, at early stages of neuronal development, expression of 5-HT6R constitutively regulates the activity of the cyclin-dependent kinase (Cdk)5 and, through this mechanism, controls cellular processes involved in circuit formation, including neuronal migration and neurite outgrowth. In addition to the Cdk5 pathway, 5-HT6R modulates a variety of key developmental targets such as Fyn, Jab1, and mammalian target of rapamycin (mTOR). Engagement of developmental pathways through 5-HT6R pharmacological manipulation has led to interesting new therapeutic perspectives in the field of psychiatric-related disorders. Indeed, 5-HT6R blockade can rescue a pathological overactivation of the mTOR pathway induced by early life insults in rodents and normalizes the associated social and episodic memory deficits. Here, we review recent evidence supporting the notion that 5-HT6R is at the interface of key developmental signaling pathways and a novel actor in the orchestration of neural circuit formation.
Collapse
Affiliation(s)
| | | | - Séverine Chaumont-Dubel
- Institut de Génomique Fonctionnelle, CNRS UMR 5203, INSERM U661, Universités Montpellier I & II, 34094 Montpellier, France
| | - Philippe Marin
- Institut de Génomique Fonctionnelle, CNRS UMR 5203, INSERM U661, Universités Montpellier I & II, 34094 Montpellier, France
| |
Collapse
|
74
|
Genetic Variations in the Serotonergic System Mediate a Combined, Weakened Response to SSRI Treatment: A Proposed Model. eNeuro 2015; 2:eN-TNC-0032-14. [PMID: 26464988 PMCID: PMC4586934 DOI: 10.1523/eneuro.0032-14.2015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 05/01/2015] [Accepted: 05/06/2015] [Indexed: 12/16/2022] Open
Abstract
Individuals with the short (S) allele in the promoter region of the serotonin transporter gene (5-HTTLPR) show a less favorable response to selective serotonin reuptake inhibitor (SSRI) treatment than individuals with the long (L) allele. Similarly, individuals with the C(-1019)G allele for the mutation found in the promoter region of the serotonin 1A receptor gene (5-HTR1A) have shown blunted responses to SSRI treatment when compared with individuals lacking this polymorphism. While these findings have been replicated across multiple studies, only two studies to date have reported data for a gene-gene interaction associated with response to SSRI treatment. Both of these studies reported a combined effect for these genotypes, with individuals homozygous for the L allele and the C allele (5-HTT(L/L)-1A(C/C)) reporting the most favorable response to SSRI treatment, and individuals homozygous for the S allele and the G allele (5-HTT(S/S)-1A(G/G)) reporting the least favorable response to SSRI treatment. Additionally, no neural mechanisms have been proposed to explain why this gene-gene interaction has been observed. To that end, this article provides a review of the relevant literature associated with these polymorphisms and proposes a feasible model that describes a genotype-dependent modulation of postsynaptic serotonin signaling associated with the 5-HTT and 5-HTR1A genes.
Collapse
|
75
|
Altieri SC, Yang H, O'Brien HJ, Redwine HM, Senturk D, Hensler JG, Andrews AM. Perinatal vs genetic programming of serotonin states associated with anxiety. Neuropsychopharmacology 2015; 40:1456-70. [PMID: 25523893 PMCID: PMC4397404 DOI: 10.1038/npp.2014.331] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 12/05/2014] [Accepted: 12/07/2014] [Indexed: 12/24/2022]
Abstract
Large numbers of women undergo antidepressant treatment during pregnancy; however, long-term consequences for their offspring remain largely unknown. Rodents exposed to serotonin transporter (SERT)-inhibiting antidepressants during development show changes in adult emotion-like behavior. These changes have been equated with behavioral alterations arising from genetic reductions in SERT. Both models are highly relevant to humans yet they vary in their time frames of SERT disruption. We find that anxiety-related behavior and, importantly, underlying serotonin neurotransmission diverge between the two models. In mice, constitutive loss of SERT causes life-long increases in anxiety-related behavior and hyperserotonemia. Conversely, early exposure to the antidepressant escitalopram (ESC; Lexapro) results in decreased anxiety-related behavior beginning in adolescence, which is associated with adult serotonin system hypofunction in the ventral hippocampus. Adult behavioral changes resulting from early fluoxetine (Prozac) exposure were different from those of ESC and, although somewhat similar to SERT deficiency, were not associated with changes in hippocampal serotonin transmission in late adulthood. These findings reveal dissimilarities in adult behavior and neurotransmission arising from developmental exposure to different widely prescribed antidepressants that are not recapitulated by genetic SERT insufficiency. Moreover, they support a pivotal role for serotonergic modulation of anxiety-related behavior.
Collapse
Affiliation(s)
- Stefanie C Altieri
- Semel Institute for Neuroscience and Human Behavior and Hatos Center for Neuropharmacology, David Geffen School of Medicine, and California NanoSystems Institute, University of California, Los Angeles, CA, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Hongyan Yang
- Semel Institute for Neuroscience and Human Behavior and Hatos Center for Neuropharmacology, David Geffen School of Medicine, and California NanoSystems Institute, University of California, Los Angeles, CA, USA
| | - Hannah J O'Brien
- Semel Institute for Neuroscience and Human Behavior and Hatos Center for Neuropharmacology, David Geffen School of Medicine, and California NanoSystems Institute, University of California, Los Angeles, CA, USA
| | - Hannah M Redwine
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Damla Senturk
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, CA, USA
| | - Julie G Hensler
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Anne M Andrews
- Semel Institute for Neuroscience and Human Behavior and Hatos Center for Neuropharmacology, David Geffen School of Medicine, and California NanoSystems Institute, University of California, Los Angeles, CA, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
76
|
Does serotonin deficit mediate susceptibility to ADHD? Neurochem Int 2015; 82:52-68. [DOI: 10.1016/j.neuint.2015.02.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 01/18/2015] [Accepted: 02/07/2015] [Indexed: 11/21/2022]
|
77
|
Clipperton-Allen AE, Page DT. Decreased aggression and increased repetitive behavior in Pten haploinsufficient mice. GENES BRAIN AND BEHAVIOR 2015; 14:145-57. [PMID: 25561290 DOI: 10.1111/gbb.12192] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 11/25/2014] [Accepted: 12/01/2014] [Indexed: 12/28/2022]
Abstract
Aggression is an aspect of social behavior that can be elevated in some individuals with autism spectrum disorder (ASD) and a concern for peers and caregivers. Mutations in Phosphatase and tensin homolog (PTEN), one of several ASD risk factors encoding negative regulators of the PI3K-Akt-mTOR pathway, have been reported in individuals with ASD and comorbid macrocephaly. We previously showed that a mouse model of Pten germline haploinsufficiency (Pten(+/-) ) has selective deficits, primarily in social behavior, along with broad overgrowth of the brain. Here, we further examine the social behavior of Pten(+/-) male mice in the resident-intruder test of aggression, using a comprehensive behavioral analysis to obtain an overall picture of the agonistic, non-agonistic and non-social behavior patterns of Pten(+/-) mice during a free interaction with a novel conspecific. Pten(+/-) male mice were involved in less aggression than their wild-type littermates. Pten(+/-) mice also performed less social investigation, including anogenital investigation and approaching and/or attending to the intruder, which is consistent with our previous finding of decreased sociability in the social approach test. In contrast to these decreases in social behaviors, Pten(+/-) mice showed increased digging. In summary, we report decreased aggression and increased repetitive behavior in Pten(+/-) mice, thus extending our characterization of this model of an ASD risk factor that features brain overgrowth and social deficits.
Collapse
|
78
|
Andolina D, Maran D, Viscomi MT, Puglisi-Allegra S. Strain-dependent variations in stress coping behavior are mediated by a 5-HT/GABA interaction within the prefrontal corticolimbic system. Int J Neuropsychopharmacol 2015; 18:pyu074. [PMID: 25522413 PMCID: PMC4360254 DOI: 10.1093/ijnp/pyu074] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Serotonin and γ-aminobutyric acid (GABA) transmission is crucial in coping strategies. METHODS Here, using mice from 2 inbred strains widely exploited in behavioral neurochemistry, we investigated whether serotonin transmission in medial prefrontal cortex and GABA in basolateral amygdala determine strain-dependent liability to stress response and differences in coping. RESULTS C57BL/6J mice displayed greater immobility in the forced swimming test, higher serotonin outflow in medial prefrontal cortex, higher GABA outflow in basolateral amygdala induced by stress, and higher serotonin 1A receptor levels in medial prefrontal cortex accompanied by lower GABAb receptor levels in basolateral amygdala than DBA/2J mice. In assessing whether serotonin in medial prefrontal cortex determines GABA functioning in response to stress and passive coping behavior in C57BL/6J and DBA/2J mice, we observed that selective prefrontal serotonin depletion in C57BL/6J and DBA/2J reduced stress-induced GABA outflow in basolateral amygdala and immobility in the forced swimming test. CONCLUSIONS These results show that strain-dependent prefrontal corticolimbic serotonin/GABA regulation determines the strain differences in stress-coping behavior in the forced swimming test and point to a role of a specific neuronal system in genetic susceptibility to stress that opens up new prospects for innovative therapies for stress disorders.
Collapse
Affiliation(s)
- Diego Andolina
- Santa Lucia Foundation, Rome, Italy (Drs Andolina, Viscomi, and Puglisi-Allegra); Dipartimento di Scienze Cliniche Applicate e Biotecnologie, Universita` degli Studi dell'Aquila, Via Vetoio, L'Aquila, Italy (Dr Andolina); Dipartimento di Psicologia and Centro 'Daniel Bovet,' Sapienza Università di Roma, Rome, Italy (Drs Maran and Puglisi-Allegra).
| | | | | | | |
Collapse
|
79
|
Tanner JA, Chenoweth MJ, Tyndale RF. Pharmacogenetics of nicotine and associated smoking behaviors. Curr Top Behav Neurosci 2015; 23:37-86. [PMID: 25655887 DOI: 10.1007/978-3-319-13665-3_3] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
This chapter summarizes genetic factors that contribute to variation in nicotine pharmacokinetics and nicotine's pharmacological action in the central nervous system (CNS), and how this in turn influences smoking behaviors. Nicotine, the major psychoactive compound in cigarette smoke, is metabolized by a number of enzymes, including CYP2A6, CYP2B6, FMOs, and UGTs, among others. Variation in the genes encoding these enzymes, in particular CYP2A6, can alter the rate of nicotine metabolism and smoking behaviors. Faster nicotine metabolism is associated with higher cigarette consumption and nicotine dependence, as well as lower quit rates. Variation in nicotine's CNS targets and downstream signaling pathways can also contribute to interindividual differences in smoking patterns. Binding of nicotine to neuronal nicotinic acetylcholine receptors (nAChRs) mediates the release of several neurotransmitters including dopamine and serotonin. Genetic variation in nAChRs, and in transporter and enzyme systems that leads to altered CNS levels of dopamine and serotonin, is associated with a number of smoking behaviors. To date, the precise mechanism underpinning many of these findings remains unknown. Considering the complex etiology of nicotine addiction, a more comprehensive approach that assesses the contribution of multiple gene variants, and their interaction with environmental factors, will likely improve personalized therapeutic approaches and increase smoking cessation rates.
Collapse
Affiliation(s)
- Julie-Anne Tanner
- Departments of Pharmacology and Toxicology and Psychiatry, University of Toronto, Toronto, ON, Canada
| | | | | |
Collapse
|
80
|
Monoamine-sensitive developmental periods impacting adult emotional and cognitive behaviors. Neuropsychopharmacology 2015; 40:88-112. [PMID: 25178408 PMCID: PMC4262911 DOI: 10.1038/npp.2014.231] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 07/30/2014] [Accepted: 08/20/2014] [Indexed: 02/07/2023]
Abstract
Development passes through sensitive periods, during which plasticity allows for genetic and environmental factors to exert indelible influence on the maturation of the organism. In the context of central nervous system development, such sensitive periods shape the formation of neurocircuits that mediate, regulate, and control behavior. This general mechanism allows for development to be guided by both the genetic blueprint as well as the environmental context. While allowing for adaptation, such sensitive periods are also vulnerability windows during which external and internal factors can confer risk to disorders by derailing otherwise resilient developmental programs. Here we review developmental periods that are sensitive to monoamine signaling and impact adult behaviors of relevance to psychiatry. Specifically, we review (1) a serotonin-sensitive period that impacts sensory system development, (2) a serotonin-sensitive period that impacts cognition, anxiety- and depression-related behaviors, and (3) a dopamine- and serotonin-sensitive period affecting aggression, impulsivity and behavioral response to psychostimulants. We discuss preclinical data to provide mechanistic insight, as well as epidemiological and clinical data to point out translational relevance. The field of translational developmental neuroscience has progressed exponentially providing solid conceptual advances and unprecedented mechanistic insight. With such knowledge at hand and important methodological innovation ongoing, the field is poised for breakthroughs elucidating the developmental origins of neuropsychiatric disorders, and thus understanding pathophysiology. Such knowledge of sensitive periods that determine the developmental trajectory of complex behaviors is a necessary step towards improving prevention and treatment approaches for neuropsychiatric disorders.
Collapse
|
81
|
Voigt JP, Fink H. Serotonin controlling feeding and satiety. Behav Brain Res 2015; 277:14-31. [PMID: 25217810 DOI: 10.1016/j.bbr.2014.08.065] [Citation(s) in RCA: 192] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 08/14/2014] [Accepted: 08/19/2014] [Indexed: 02/06/2023]
|
82
|
Kim J, Rim YS, Chen H, Cao HH, Nakatsuka N, Hinton HL, Zhao C, Andrews AM, Yang Y, Weiss PS. Fabrication of High-Performance Ultrathin In2O3 Film Field-Effect Transistors and Biosensors Using Chemical Lift-Off Lithography. ACS NANO 2015; 9:4572-82. [PMID: 25798751 DOI: 10.1021/acsnano.5b01211] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
We demonstrate straightforward fabrication of highly sensitive biosensor arrays based on field-effect transistors, using an efficient high-throughput, large-area patterning process. Chemical lift-off lithography is used to construct field-effect transistor arrays with high spatial precision suitable for the fabrication of both micrometer- and nanometer-scale devices. Sol-gel processing is used to deposit ultrathin (∼4 nm) In2O3 films as semiconducting channel layers. The aqueous sol-gel process produces uniform In2O3 coatings with thicknesses of a few nanometers over large areas through simple spin-coating, and only low-temperature thermal annealing of the coatings is required. The ultrathin In2O3 enables construction of highly sensitive and selective biosensors through immobilization of specific aptamers to the channel surface; the ability to detect subnanomolar concentrations of dopamine is demonstrated.
Collapse
Affiliation(s)
- Jaemyung Kim
- †California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
- ‡Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - You Seung Rim
- †California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
- §Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Huajun Chen
- †California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
- §Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Huan H Cao
- †California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
- ‡Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Nako Nakatsuka
- †California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
- ‡Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Hannah L Hinton
- †California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
- ‡Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Chuanzhen Zhao
- †California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
- ‡Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
- ⊥Department of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Anne M Andrews
- †California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
- ‡Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
- ∥Department of Psychiatry and Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Yang Yang
- †California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
- §Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Paul S Weiss
- †California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
- ‡Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
- §Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| |
Collapse
|
83
|
Developmental alterations in anxiety and cognitive behavior in serotonin transporter mutant mice. Psychopharmacology (Berl) 2014; 231:4119-33. [PMID: 24728652 DOI: 10.1007/s00213-014-3554-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 03/17/2014] [Indexed: 10/25/2022]
Abstract
RATIONALE A promoter variant of the serotonin transporter (SERT) gene is known to affect emotional and cognitive regulation. In particular, the "short" allelic variant is implicated in the etiology of multiple neuropsychiatric disorders. Heterozygous (SERT(+/-)) and homozygous (SERT(-/-)) SERT mutant mice are valuable tools for understanding the mechanisms of altered SERT levels. Although these genetic effects are well investigated in adulthood, the developmental trajectory of altered SERT levels for behavior has not been investigated. OBJECTIVES We assessed anxiety-like and cognitive behaviors in SERT mutant mice in early adolescence and adulthood to examine the developmental consequences of reduced SERT levels. Spine density of pyramidal neurons was also measured in corticolimbic brain regions. RESULTS Adult SERT(-/-) mice exhibited increased anxiety-like behavior, but these differences were not observed in early adolescent SERT(-/-) mice. Conversely, SERT(+/-) and SERT(-/-) mice did display higher spontaneous alternation during early adolescence and adulthood. SERT(+/-) and SERT(-/-) also exhibited greater neuronal spine densities in the orbitofrontal but not the medial prefrontal cortices. Adult SERT(-/-) mice also showed an increased spine density in the basolateral amygdala. CONCLUSIONS Developmental alterations of the serotonergic system caused by genetic inactivation of SERT can have different influences on anxiety-like and cognitive behaviors through early adolescence into adulthood, which may be associated with changes of spine density in the prefrontal cortex and amygdala. The altered maturation of serotonergic systems may lead to specific age-related vulnerabilities to psychopathologies that develop during adolescence.
Collapse
|
84
|
Serotonergic versus nonserotonergic dorsal raphe projection neurons: differential participation in reward circuitry. Cell Rep 2014; 8:1857-1869. [PMID: 25242321 DOI: 10.1016/j.celrep.2014.08.037] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 05/22/2014] [Accepted: 08/15/2014] [Indexed: 12/20/2022] Open
Abstract
The dorsal raphe nucleus (DRN) contains the largest group of serotonin-producing neurons in the brain and projects to regions controlling reward. Although pharmacological studies suggest that serotonin inhibits reward seeking, electrical stimulation of the DRN strongly reinforces instrumental behavior. Here, we provide a targeted assessment of the behavioral, anatomical, and electrophysiological contributions of serotonergic and nonserotonergic DRN neurons to reward processes. To explore DRN heterogeneity, we used a simultaneous two-vector knockout/optogenetic stimulation strategy, as well as cre-induced and cre-silenced vectors in several cre-expressing transgenic mouse lines. We found that the DRN is capable of reinforcing behavior primarily via nonserotonergic neurons, for which the main projection target is the ventral tegmental area (VTA). Furthermore, these nonserotonergic projections provide glutamatergic excitation of VTA dopamine neurons and account for a large majority of the DRN-VTA pathway. These findings help to resolve apparent discrepancies between the roles of serotonin versus the DRN in behavioral reinforcement.
Collapse
|
85
|
Ethanol effects on multiple fixed-interval, fixed-ratio responding in mice with deletions of the serotonin transporter gene. Behav Pharmacol 2014; 25:92-5. [PMID: 24247279 DOI: 10.1097/fbp.0000000000000011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Serotonin transporter knockout (KO) mice self-administer less ethanol than either heterozygous or wild-type mice; however, the mechanistic basis for this difference remains unclear. Here we examine the possibility that ethanol more readily decreases responding in KO mice, thereby limiting ethanol self-administration. To examine whether KO mice were more sensitive to the response-decreasing effects of ethanol, we administered ethanol (0.2-3.2 g/kg) to mice responding under a multiple fixed-ratio 30-response, fixed-interval 300-s schedule of milk presentation. Ethanol decreased responding similarly in all three genotypes. Fixed-ratio responding tended to be decreased at lower doses than fixed-interval responding. The decreased level of ethanol self-administration in serotonin transporter KO mice is not explained by an increased sensitivity to the response-decreasing effects of ethanol in KO mice, as sensitivity to the response-decreasing effects of ethanol was similar in the KO, heterozygous, and wild-type mice.
Collapse
|
86
|
Ramadan E, Blanchard H, Cheon Y, Fox MA, Chang L, Chen M, Ma K, Rapoport SI, Basselin M. Transient postnatal fluoxetine leads to decreased brain arachidonic acid metabolism and cytochrome P450 4A in adult mice. Prostaglandins Leukot Essent Fatty Acids 2014; 90:191-7. [PMID: 24529827 PMCID: PMC3981912 DOI: 10.1016/j.plefa.2014.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 01/19/2014] [Accepted: 01/21/2014] [Indexed: 12/11/2022]
Abstract
Fetal and perinatal exposure to selective serotonin (5-HT) reuptake inhibitors (SSRIs) has been reported to alter childhood behavior, while transient early exposure in rodents is reported to alter their behavior and decrease brain extracellular 5-HT in adulthood. Since 5-HT2A/2C receptor-mediated neurotransmission can involve G-protein coupled activation of cytosolic phospholipase A2 (cPLA2), releasing arachidonic acid (ARA) from synaptic membrane phospholipid, we hypothesized that transient postnatal exposure to fluoxetine would alter brain ARA metabolism in adult mice. Brain ARA incorporation coefficients k* and rates Jin were quantitatively imaged following intravenous [1-(14)C]ARA infusion of unanesthetized adult mice that had been injected daily with fluoxetine (10mg/kg i.p.) or saline during postnatal days P4-P21. Expression of brain ARA metabolic enzymes and other relevant markers also was measured. On neuroimaging, k* and Jin was decreased widely in early fluoxetine- compared to saline-treated adult mice. Of the enzymes measured, cPLA2 activity was unchanged, while Ca(2+)-independent iPLA2 activity was increased. There was a significant 74% reduced protein level of cytochrome P450 (CYP) 4A, which can convert ARA to 20-HETE. Reduced brain ARA metabolism in adult mice transiently exposed to postnatal fluoxetine, and a 74% reduction in CYP4A protein, suggest long-term effects independent of drug presence in brain ARA metabolism, and in CYP4A metabolites. These changes might contribute to reported altered behavior following early SSRI in rodents.
Collapse
Affiliation(s)
- Epolia Ramadan
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Helene Blanchard
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Yewon Cheon
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Meredith A Fox
- Laboratory of Clinical Science, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lisa Chang
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Mei Chen
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Kaizong Ma
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Stanley I Rapoport
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Mireille Basselin
- Brain Physiology and Metabolism Section, Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
87
|
Birbeck JA, Khalid M, Mathews TA. Potentiated striatal dopamine release leads to hyperdopaminergia in female brain-derived neurotrophic factor heterozygous mice. ACS Chem Neurosci 2014; 5:275-81. [PMID: 24517838 DOI: 10.1021/cn400157b] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The goal of this study was to determine whether a reduction in brain-derived neurotrophic factor (BDNF) levels in female mice leads to dopaminergic system dysregulation. Through a series of in vivo brain microdialysis and slice voltammetry experiments, we discerned that female BDNF heterozygous (BDNF(+/-)) mice are hyperdopaminergic, similar to their male BDNF(+/-) counterparts. Zero-net flux microdialysis results showed that female BDNF(+/-) mice had increased striatal extracellular dopamine levels, while stimulated regional release by high potassium concentrations potentiated dopamine release through vesicular-mediated depolarization. Using the complementary technique of fast scan cyclic voltammetry, electrical stimulation evoked greater dopamine release in the female BDNF(+/-) mice, whereas dopamine uptake remained unchanged relative to that of female wildtype mice. Following psychostimulant methamphetamine administration, female BDNF(+/-) mice showed potentiated dopamine release compared to their wildtype counterparts. Taken together, these dopamine release impairments in female mice appear to result in a hyperdopaminergic phenotype without concomitant alterations in dopamine uptake.
Collapse
Affiliation(s)
- Johnna A. Birbeck
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| | - Madiha Khalid
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| | - Tiffany A. Mathews
- Department of Chemistry, Wayne State University, Detroit, Michigan 48202, United States
| |
Collapse
|
88
|
Schröder CM, Primeau MM, Hallmayer JF, Lazzeroni LC, Hubbard JT, O’Hara R. Serotonin transporter polymorphism is associated with increased apnea-hypopnea index in older adults. Int J Geriatr Psychiatry 2014; 29:227-35. [PMID: 23754303 PMCID: PMC3883911 DOI: 10.1002/gps.3994] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 05/02/2013] [Indexed: 12/22/2022]
Abstract
RATIONALE A functional polymorphism of the serotonin transporter gene (5-HTTLPR) has previously been related to upper airway pathology, but its contribution to obstructive sleep apnea (OSA), a highly prevalent sleep disorder in older adults, remains unclear. OBJECTIVES We aimed to investigate the relationship between apnea-hypopnea index (AHI) and genetic variations in the promoter region of the 5-HTTLPR in older adults. METHODS DNA samples from 94 community-dwelling older adults (57% female, mean age 72 ± 8) were genotyped for the 5-HTTLPR polymorphism. All participants were assessed in their homes with full ambulatory polysomnography in order to determine AHI and related parameters such as hypoxia, sleep fragmentation, and self-reported daytime sleepiness. RESULTS The 5-HTT l allele was significantly associated with AHI (p = 0.019), with l allele carriers displaying a higher AHI than s allele homozygotes. A single allele change in 5-HTTLPR genotype from s to l resulted in an increase of AHI by 4.46 per hour of sleep (95% CI, 0.75-8.17). The l allele was also associated with increased time during sleep spent at oxygen saturation levels below 90% (p = 0.014). CONCLUSIONS The observed significant association between the 5-HTTLPR l allele and severity of OSA in older adults suggests that the l allele may be important to consider when assessing for OSA in this age group. This association may also explain some of the observed variability among serotonergic pharmacological treatment studies for OSA, and 5-HTT genotype status may have to be taken into account in future therapeutic trials involving serotonergic agents.
Collapse
Affiliation(s)
- Carmen M. Schröder
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 401 Quarry Road, Stanford University, Stanford, CA 94305-5550, USA
- Department of Psychiatry and Behavioral Sciences, Strasbourg University Hospital and Strasbourg University, 1 place de l’hôpital, 67000 Strasbourg, France
- CNRS UPR 3212, Institute for Cellular and Integrative Neuroscience, Strasbourg, France
- University Sleep Clinic, Department of Neurology, Strasbourg University Hospital, Strasbourg, France
| | - Michelle M. Primeau
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 401 Quarry Road, Stanford University, Stanford, CA 94305-5550, USA
| | - Joachim F. Hallmayer
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 401 Quarry Road, Stanford University, Stanford, CA 94305-5550, USA
| | - Laura C. Lazzeroni
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 401 Quarry Road, Stanford University, Stanford, CA 94305-5550, USA
| | - Jeffrey T. Hubbard
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 401 Quarry Road, Stanford University, Stanford, CA 94305-5550, USA
- CNRS UPR 3212, Institute for Cellular and Integrative Neuroscience, Strasbourg, France
- University Sleep Clinic, Department of Neurology, Strasbourg University Hospital, Strasbourg, France
| | - Ruth O’Hara
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 401 Quarry Road, Stanford University, Stanford, CA 94305-5550, USA
| |
Collapse
|
89
|
Lasting neurobehavioral abnormalities in rats after neonatal activation of serotonin 1A and 1B receptors: possible mechanisms for serotonin dysfunction in autistic spectrum disorders. Psychopharmacology (Berl) 2014; 231:1191-200. [PMID: 23975037 PMCID: PMC3933458 DOI: 10.1007/s00213-013-3242-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 08/06/2013] [Indexed: 10/26/2022]
Abstract
RATIONALE Perinatal exposure of rats to selective serotonin reuptake inhibitors (SSRIs) produces sensory and social abnormalities paralleling those seen in autistic spectrum disorders (ASDs). However, the possible mechanism(s) by which this exposure produces behavioral abnormalities is unclear. OBJECTIVE We hypothesized that the lasting effects of neonatal SSRI exposure are a consequence of abnormal stimulation of 5-HT1A and/or 5-HT1B receptors during brain development. We examined whether such stimulation would result in lasting sensory and social deficits in rats in a manner similar to SSRIs using both direct agonist stimulation of receptors as well as selective antagonism of these receptors during SSRI exposure. METHODS Male and female rat pups were treated from postnatal days 8 to 21. In Experiment 1, pups received citalopram (20 mg/kg/day), saline, (±)-8-hydroxy-dipropylaminotetralin hydrobromide (8-OH-DPAT; 0.5 mg/kg/day) or 7-trifluoromethyl-4(4-methyl-1-piperazinyl)-pyrrolo[1,2-a]-quinoxaline dimaleate (CGS-12066B; 10 mg/kg/day). In Experiment 2, a separate cohort of pups received citalopram (20 mg/kg/day), or saline which was combined with either N-[2-[4-(2-methoxyphenyl)-1-piperazinyl]ethyl]-N-2-pyridinylcyclo-hexanecarboxamide maleate (WAY-100635; 0.6 mg/kg/day) or N-[4-methoxy-3-(4-methyl-1-piperazinyl)phenyl]-2'-methyl-4'-(5-methyl-1,2,4-oxadiazol-3-yl)-1-1'-biphenyl-4-carboxamide (GR-127935; 6 mg/kg/day) or vehicle. Rats were then tested in paradigms designed to assess sensory and social response behaviors at different time points during development. RESULTS Direct and indirect neonatal stimulation of 5-HT1A or 5-HT1B receptors disrupts sensory processing, produces neophobia, increases stereotypic activity, and impairs social interactions in manner analogous to that observed in ASD. CONCLUSION Increased stimulation of 5-HT1A and 5-HT1B receptors plays a significant role in the production of lasting social and sensory deficits in adult animals exposed as neonates to SSRIs.
Collapse
|
90
|
Overexpression of gastric leptin precedes adipocyte leptin during high-fat diet and is linked to 5HT-containing enterochromaffin cells. Int J Obes (Lond) 2014; 38:1357-64. [PMID: 24468700 DOI: 10.1038/ijo.2014.14] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 12/20/2013] [Accepted: 01/17/2014] [Indexed: 12/20/2022]
|
91
|
Fox MA, Panessiti MG, Moya PR, Tolliver TJ, Chen K, Shih JC, Murphy DL. Mutations in monoamine oxidase (MAO) genes in mice lead to hypersensitivity to serotonin-enhancing drugs: implications for drug side effects in humans. THE PHARMACOGENOMICS JOURNAL 2013; 13:551-7. [PMID: 22964922 PMCID: PMC3562558 DOI: 10.1038/tpj.2012.35] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Revised: 07/18/2012] [Accepted: 07/23/2012] [Indexed: 12/16/2022]
Abstract
A possible side effect of serotonin-enhancing drugs is the serotonin syndrome, which can be lethal. Here we examined possible hypersensitivity to two such drugs, the serotonin precursor 5-hydroxy-L-tryptophan (5-HTP) and the atypical opioid tramadol, in mice lacking the genes for both monoamine oxidase A (MAOA) and MAOB. MAOA/B-knockout (KO) mice displayed baseline serotonin syndrome behaviors, and these behavioral responses were highly exaggerated following 5-HTP or tramadol versus baseline and wild-type (WT) littermates. Compared with MAOA/B-WT mice, baseline tissue serotonin levels were increased ∼2.6-3.9-fold in MAOA/B-KO mice. Following 5-HTP, serotonin levels were further increased ∼4.5-6.2-fold in MAOA/B-KO mice. These exaggerated responses are in line with the exaggerated responses following serotonin-enhancing drugs that we previously observed in mice lacking the serotonin transporter (SERT). These findings provide a second genetic mouse model suggestive of possible human vulnerability to the serotonin syndrome in individuals with lesser-expressing MAO or SERT polymorphisms that confer serotonergic system changes.
Collapse
Affiliation(s)
- MA Fox
- Laboratory of Clinical Science (LCS), National Institute of Mental Health, NIH, Bethesda, MD, USA
| | - MG Panessiti
- Laboratory of Clinical Science (LCS), National Institute of Mental Health, NIH, Bethesda, MD, USA
| | - PR Moya
- Laboratory of Clinical Science (LCS), National Institute of Mental Health, NIH, Bethesda, MD, USA
| | - TJ Tolliver
- Laboratory of Clinical Science (LCS), National Institute of Mental Health, NIH, Bethesda, MD, USA
| | - K Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - JC Shih
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
- Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - DL Murphy
- Laboratory of Clinical Science (LCS), National Institute of Mental Health, NIH, Bethesda, MD, USA
| |
Collapse
|
92
|
Prefrontal/amygdalar system determines stress coping behavior through 5-HT/GABA connection. Neuropsychopharmacology 2013; 38:2057-67. [PMID: 23636466 PMCID: PMC3746690 DOI: 10.1038/npp.2013.107] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Revised: 04/20/2013] [Accepted: 04/22/2013] [Indexed: 11/08/2022]
Abstract
Coping is defined as the behavioral and physiological effort made to master stressful situations. The ability to cope with stress leads either to healthy or to pathogenic outcomes. The medial prefrontal cortex (mpFC) and amygdala are acknowledged as having a major role in stress-related behaviors, and mpFC has a critical role in the regulation of amygdala-mediated arousal in response to emotionally salient stimuli. Prefrontal cortical serotonin (5-hydroxytryptamine (5-HT)) is involved in corticolimbic circuitry, and GABA has a major role in amygdala functioning. Here, using mice, it was assessed whether amygdalar GABA regulation by prefrontal 5-HT is involved in processing stressful experiences and in determining coping outcomes. First (experiment 1), bilateral selective 5-HT depletion in mpFC of mice reduced GABA release induced by stress in basolateral amygdala (BLA) and passive coping in the Forced Swimming Test (FST) (experiment 2). Moreover, prefrontal-amygdala disconnection procedure that combined a selective unilateral 5-HT depletion of mpFC and infusion of an inhibitor of GABA synthesis into the contralateral BLA, thereby to disrupt prefrontal-amygdalar serial connectivity bilaterally, showed that disconnection selectively decreases immobility in the FST. These results point to prefrontal/amygdala connectivity mediated by 5-HT and GABA transmission as a critical neural mechanism in stress-induced behavior.
Collapse
|
93
|
Lamb RJ, Daws LC. Ethanol self-administration in serotonin transporter knockout mice: unconstrained demand and elasticity. GENES BRAIN AND BEHAVIOR 2013; 12:741-7. [PMID: 23927813 DOI: 10.1111/gbb.12068] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 06/03/2013] [Accepted: 08/02/2013] [Indexed: 10/26/2022]
Abstract
Low serotonin function is associated with alcoholism, leading to speculation that increasing serotonin function could decrease ethanol consumption. Mice with one or two deletions of the serotonin transporter (SERT) gene have increased extracellular serotonin. To examine the relationship between SERT genotype and motivation for alcohol, we compared ethanol self-administration in mice with zero (knockout, KO), one (HET) or two copies (WT) of the SERT gene. All three genotypes learned to self-administer ethanol. The SSRI, fluvoxamine, decreased responding for ethanol in the HET and WT, but not the KO mice. When tested under a progressive ratio schedule, KO mice had lower breakpoints than HET or WT. As work requirements were increased across sessions, behavioral economic analysis of ethanol self-administration indicated that the decreased breakpoint in KO as compared to HET or WT mice was a result of lower levels of unconstrained demand, rather than differences in elasticity, i.e. the proportional decreases in ethanol earned with increasing work requirements were similar across genotypes. The difference in unconstrained demand was unlikely to result from motor or general motivational factors, as both WT and KO mice responded at high levels for a 50% condensed milk solution. As elasticity is hypothesized to measure essential value, these results indicate that KO value ethanol similarly to WT or HET mice despite having lower break points for ethanol.
Collapse
Affiliation(s)
- R J Lamb
- Department of Psychiatry; Department of Pharmacology
| | | |
Collapse
|
94
|
Araragi N, Mlinar B, Baccini G, Gutknecht L, Lesch KP, Corradetti R. Conservation of 5-HT1A receptor-mediated autoinhibition of serotonin (5-HT) neurons in mice with altered 5-HT homeostasis. Front Pharmacol 2013; 4:97. [PMID: 23935583 PMCID: PMC3731744 DOI: 10.3389/fphar.2013.00097] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 07/17/2013] [Indexed: 12/23/2022] Open
Abstract
Firing activity of serotonin (5-HT) neurons in the dorsal raphe nucleus (DRN) is controlled by inhibitory somatodendritic 5-HT1A autoreceptors. This autoinhibitory mechanism is implicated in the etiology of disorders of emotion regulation, such as anxiety disorders and depression, as well as in the mechanism of antidepressant action. Here, we investigated how persistent alterations in brain 5-HT availability affect autoinhibition in two genetically modified mouse models lacking critical mediators of serotonergic transmission: 5-HT transporter knockout (Sert-/-) and tryptophan hydroxylase-2 knockout (Tph2-/-) mice. The degree of autoinhibition was assessed by loose-seal cell-attached recording in DRN slices. First, application of the 5-HT1A-selective agonist R(+)-8-hydroxy-2-(di-n-propylamino)tetralin showed mild sensitization and marked desensitization of 5-HT1A receptors in Tph2-/- mice and Sert-/- mice, respectively. While 5-HT neurons from Tph2-/- mice did not display autoinhibition in response to L-tryptophan, autoinhibition of these neurons was unaltered in Sert-/- mice despite marked desensitization of their 5-HT1A autoreceptors. When the Tph2-dependent 5-HT synthesis step was bypassed by application of 5-hydroxy-L-tryptophan (5-HTP), neurons from both Tph2-/- and Sert-/- mice decreased their firing rates at significantly lower concentrations of 5-HTP compared to wildtype controls. Our findings demonstrate that, as opposed to the prevalent view, sensitivity of somatodendritic 5-HT1A receptors does not predict the magnitude of 5-HT neuron autoinhibition. Changes in 5-HT1A receptor sensitivity may rather be seen as an adaptive mechanism to keep autoinhibition functioning in response to extremely altered levels of extracellular 5-HT resulting from targeted inactivation of mediators of serotonergic signaling.
Collapse
Affiliation(s)
- Naozumi Araragi
- Division of Molecular Psychiatry, Laboratory of Translational Neuroscience, Department of Psychiatry, Psychosomatics, and Psychotherapy, University of Wuerzburg Wuerzburg, Germany
| | | | | | | | | | | |
Collapse
|
95
|
Birbeck JA, Mathews TA. Simultaneous Detection of Monoamine and Purine Molecules Using High-Performance Liquid Chromatography with a Boron-Doped Diamond Electrode. Anal Chem 2013; 85:7398-404. [DOI: 10.1021/ac4013144] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Johnna A. Birbeck
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit,
Michigan 48202, United States
| | - Tiffany A. Mathews
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit,
Michigan 48202, United States
| |
Collapse
|
96
|
Kinast K, Peeters D, Kolk SM, Schubert D, Homberg JR. Genetic and pharmacological manipulations of the serotonergic system in early life: neurodevelopmental underpinnings of autism-related behavior. Front Cell Neurosci 2013; 7:72. [PMID: 23781172 PMCID: PMC3679613 DOI: 10.3389/fncel.2013.00072] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Accepted: 04/30/2013] [Indexed: 12/28/2022] Open
Abstract
Serotonin, in its function as neurotransmitter, is well-known for its role in depression, autism and other neuropsychiatric disorders, however, less known as a neurodevelopmental factor. The serotonergic system is one of the earliest to develop during embryogenesis and early changes in serotonin levels can have large consequences for the correct development of specific brain areas. The regulation and functioning of serotonin is influenced by genetic risk factors, such as the serotonin transporter polymorphism in humans. This polymorphism is associated with anxiety-related symptoms, changes in social behavior, and cortical gray and white matter changes also seen in patients suffering from autism spectrum disorders (ASD). The human polymorphism can be mimicked by the knockout of the serotonin transporter in rodents, which are as a model system therefore vital to explore the precise neurobiological mechanisms. Moreover, there are pharmacological challenges influencing serotonin in early life, like prenatal/neonatal exposure to selective serotonin reuptake inhibitors (SSRI) in depressed pregnant women. There is accumulating evidence that this dysregulation of serotonin during critical phases of brain development can lead to ASD-related symptoms in children, and reduced social behavior and increased anxiety in rodents. Furthermore, prenatal valproic acid (VPA) exposure, a mood stabilizing drug which is also thought to interfere with serotonin levels, has the potency to induce ASD-like symptoms and to affect the development of the serotonergic system. Here, we review and compare the neurodevelopmental and behavioral consequences of serotonin transporter gene variation, and prenatal SSRI and VPA exposure in the context of ASD.
Collapse
Affiliation(s)
- Karsten Kinast
- Behavioural Neurogenetics, Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Nijmegen Medical Centre Nijmegen, Netherlands
| | | | | | | | | |
Collapse
|
97
|
Olivier JDA, Vinkers CH, Olivier B. The role of the serotonergic and GABA system in translational approaches in drug discovery for anxiety disorders. Front Pharmacol 2013; 4:74. [PMID: 23781201 PMCID: PMC3677985 DOI: 10.3389/fphar.2013.00074] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 05/21/2013] [Indexed: 12/13/2022] Open
Abstract
There is ample evidence that genetic factors play an important role in anxiety disorders. In support, human genome-wide association studies have implicated several novel candidate genes. However, illumination of such genetic factors involved in anxiety disorders has not resulted in novel drugs over the past decades. A complicating factor is the heterogeneous classification of anxiety disorders in the Diagnostic and Statistical Manual of Mental Disorders (DSM-IV-TR) and diverging operationalization of anxiety used in preclinical and clinical studies. Currently, there is an increasing focus on the gene × environment (G × E) interaction in anxiety as genes do not operate in isolation and environmental factors have been found to significantly contribute to the development of anxiety disorders in at-risk individuals. Nevertheless, extensive research on G × E mechanisms in anxiety has not resulted in major breakthroughs in drug discovery. Modification of individual genes in rodent models has enabled the specific study of anxiety in preclinical studies. In this context, two extensively studied neurotransmitters involved in anxiety are the gamma-aminobutyric acid (GABA) and 5-HT (5-hydroxytryptamine) system. In this review, we illustrate the complex interplay between genes and environment in anxiety processes by reviewing preclinical and clinical studies on the serotonin transporter (5-HTT), 5-HT1A receptor, 5-HT2 receptor, and GABAA receptor. Even though targets from the serotonin and GABA system have yielded drugs with known anxiolytic efficacy, the relation between the genetic background of these targets and anxiety symptoms and development of anxiety disorders is largely unknown. The aim of this review is to show the vast complexity of genetic and environmental factors in anxiety disorders. In light of the difficulty with which common genetic variants are identified in anxiety disorders, animal models with translational validity may aid in elucidating the neurobiological background of these genes and their possible role in anxiety. We argue that, in addition to human genetic studies, translational models are essential to map anxiety-related genes and to enhance our understanding of anxiety disorders in order to develop potentially novel treatment strategies.
Collapse
Affiliation(s)
- Jocelien D A Olivier
- Department of, Women's and Children's Health, Uppsala University Uppsala, Sweden ; Center for Gender Medicine, Karolinska Institutet Stockholm, Sweden
| | | | | |
Collapse
|
98
|
Fitzgerald PJ. Black bile: Are elevated monoamines an etiological factor in some cases of major depression? Med Hypotheses 2013; 80:823-6. [DOI: 10.1016/j.mehy.2013.03.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 03/17/2013] [Indexed: 10/27/2022]
|
99
|
Yang H, Thompson A, McIntosh BJ, Altieri SC, Andrews AM. Physiologically relevant changes in serotonin resolved by fast microdialysis. ACS Chem Neurosci 2013; 4:790-8. [PMID: 23614776 PMCID: PMC3656759 DOI: 10.1021/cn400072f] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Accepted: 04/03/2013] [Indexed: 01/17/2023] Open
Abstract
Online microdialysis is a sampling and detection method that enables continuous interrogation of extracellular molecules in freely moving subjects under behaviorally relevant conditions. A majority of recent publications using brain microdialysis in rodents report sample collection times of 20-30 min. These long sampling times are due, in part, to limitations in the detection sensitivity of high performance liquid chromatography (HPLC). By optimizing separation and detection conditions, we decreased the retention time of serotonin to 2.5 min and the detection threshold to 0.8 fmol. Sampling times were consequently reduced from 20 to 3 min per sample for online detection of serotonin (and dopamine) in brain dialysates using a commercial HPLC system. We developed a strategy to collect and to analyze dialysate samples continuously from two animals in tandem using the same instrument. Improvements in temporal resolution enabled elucidation of rapid changes in extracellular serotonin levels associated with mild stress and circadian rhythms. These dynamics would be difficult or impossible to differentiate using conventional microdialysis sampling rates.
Collapse
Affiliation(s)
- Hongyan Yang
- Semel Institute for Neuroscience & Human
Behavior and Hatos Center for Neuropharmacology, David Geffen School
of Medicine, Department of Chemistry and Biochemistry, and California NanoSystems Institute, University of California, Los Angeles,
California, United States
| | - Andrew
B. Thompson
- Semel Institute for Neuroscience & Human
Behavior and Hatos Center for Neuropharmacology, David Geffen School
of Medicine, Department of Chemistry and Biochemistry, and California NanoSystems Institute, University of California, Los Angeles,
California, United States
| | | | - Stefanie C. Altieri
- Semel Institute for Neuroscience & Human
Behavior and Hatos Center for Neuropharmacology, David Geffen School
of Medicine, Department of Chemistry and Biochemistry, and California NanoSystems Institute, University of California, Los Angeles,
California, United States
| | - Anne M. Andrews
- Semel Institute for Neuroscience & Human
Behavior and Hatos Center for Neuropharmacology, David Geffen School
of Medicine, Department of Chemistry and Biochemistry, and California NanoSystems Institute, University of California, Los Angeles,
California, United States
| |
Collapse
|
100
|
Jennings KA. A comparison of the subsecond dynamics of neurotransmission of dopamine and serotonin. ACS Chem Neurosci 2013; 4:704-14. [PMID: 23627553 DOI: 10.1021/cn4000605] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The neuromodulators dopamine (DA) and serotonin (5-hydroxytryptamine; 5-HT) are similar in a number of ways. Both monoamines can act by volume transmission at metabotropic receptors to modulate synaptic transmission in brain circuits. Presynaptic regulation of 5-HT and DA is governed by parallel processes, and behaviorally, both exert control over emotional processing. However, differences are also apparent: more than twice as many 5-HT receptor subtypes mediate postsynaptic effects than DA receptors and different presynaptic regulation is also emerging. Monoamines are amenable to real-time electrochemical detection using fast scan cyclic voltammetry (FSCV), which allows resolution of the subsecond dynamics of release and reuptake in response to a single action potential. This approach has greatly enriched understanding of DA transmission and has facilitated an integrated view of how DA mediates behavioral control. However, technical challenges are associated with FSCV measurement of 5-HT and understanding of 5-HT transmission at subsecond resolution has not advanced at the same rate. As a result, how the actions of 5-HT at the level of the synapse translate into behavior is poorly understood. Recent technical advances may aid the study of 5-HT in real-time. It is timely, therefore, to compare and contrast what is currently understood of the subsecond characteristics of transmission for DA and 5-HT. In doing so, a number of areas are highlighted as being worthy of exploration for 5-HT.
Collapse
Affiliation(s)
- Katie A. Jennings
- Department of Physiology, Anatomy and Genetics, Oxford University, South Parks Road, Oxford, U.K. OX1
3PT
| |
Collapse
|