1
|
Dong J, Wei X, Huang Z, Tian J, Zhang W. Age-related changes of dopamine D1 and D2 receptors expression in parvalbumin-positive cells of the orbitofrontal and prelimbic cortices of mice. Front Neurosci 2024; 18:1364067. [PMID: 38903598 PMCID: PMC11187244 DOI: 10.3389/fnins.2024.1364067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 05/28/2024] [Indexed: 06/22/2024] Open
Abstract
Dopamine (DA) plays a pivotal role in reward processing, cognitive functions, and emotional regulation. The prefrontal cortex (PFC) is a critical brain region for these processes. Parvalbumin-positive (PV+) neurons are one of the major classes of inhibitory GABAergic neurons in the cortex, they modulate the activity of neighboring neurons, influencing various brain functions. While DA receptor expression exhibits age-related changes, the age-related changes of these receptors in PV+ neurons, especially in the PFC, remain unclear. To address this, we investigated the expression of DA D1 (D1R) and D2 (D2R) receptors in PV+ neurons within the orbitofrontal (OFC) and prelimbic (PrL) cortices at different postnatal ages (P28, P42, P56, and P365). We found that the expression of D1R and D2R in PV+ neurons showed both age- and region-related changes. PV+ neurons in the OFC expressed a higher abundance of D1 than those in the PrL, and those neurons in the OFC also showed higher co-expression of D1R and D2R than those in the PrL. In the OFC and PrL, D1R in PV+ neurons increased from P28 and reached a plateau at P42, then receded to express at P365. Meanwhile, D2R did not show significant age-related changes between the two regions except at P56. These results showed dopamine receptors in the prefrontal cortex exhibit age- and region-specific changes, which may contribute to the difference of these brain regions in reward-related brain functions.
Collapse
Affiliation(s)
- Jihui Dong
- Department of Neurobiology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, China
- Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
| | - Xiaoyan Wei
- Department of Neurobiology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, China
- Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
| | - Ziran Huang
- Department of Neurobiology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, China
- Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
| | - Jing Tian
- Department of Neurobiology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, China
- Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
| | - Wen Zhang
- Department of Neurobiology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, China
- Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
| |
Collapse
|
2
|
Caldwell M, Mendoza JC, Jiang XYZ, Alarcon C, Ayo-Jibunoh V, Louis S, Maronna D, Darwish R, Tomaio J, Mingote S, Yetnikoff L. Reorganization of dopamine circuitry in the anterior corpus callosum between early adolescence and adulthood in the mouse. Eur J Neurosci 2024; 59:2535-2548. [PMID: 38720367 DOI: 10.1111/ejn.16385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 04/13/2024] [Accepted: 04/22/2024] [Indexed: 05/22/2024]
Abstract
The maturation of forebrain dopamine circuitry occurs over multiple developmental periods, extending from early postnatal life until adulthood, with the precise timing of maturation defined by the target region. We recently demonstrated in the adult mouse brain that axon terminals arising from midbrain dopamine neurons innervate the anterior corpus callosum and that oligodendrocyte lineage cells in this white matter tract express dopamine receptor transcripts. Whether corpus callosal dopamine circuitry undergoes maturational changes between early adolescence and adulthood is unknown but may be relevant to understanding the dramatic micro- and macro-anatomical changes that occur in the corpus callosum of multiple species during early adolescence, including in the degree of myelination. Using quantitative neuroanatomy, we show that dopamine innervation in the forceps minor, but not the rostral genu, of the corpus callosum, is greater during early adolescence (P21) compared to adulthood (>P90) in wild-type mice. We further demonstrate with RNAscope that, as in the adult, Drd1 and Drd2 transcripts are expressed at higher levels in oligodendrocyte precursor cells (OPCs) and decline as these cells differentiate into oligodendrocytes. In addition, the number of OPCs that express Drd1 transcripts during early adolescence is double the number of those expressing the transcript during early adulthood. These data further implicate dopamine in axon myelination and myelin regulation. Moreover, because developmental (activity-independent) myelination peaks during early adolescence, with experience-dependent (activity-dependent) myelination greatest during early adulthood, our data suggest that potential roles of dopamine on callosal myelination shift between early adolescence and adulthood, from a developmental role to an experience-dependent role.
Collapse
Affiliation(s)
- Megan Caldwell
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, New York, NY, USA
| | - Josue Criollo Mendoza
- Department of Biology, College of Staten Island, City University of New York, Staten Island, NY, USA
| | - Xin Yan Zhu Jiang
- Department of Biology, College of Staten Island, City University of New York, Staten Island, NY, USA
| | - Colin Alarcon
- Department of Psychology, College of Staten Island, City University of New York, Staten Island, NY, USA
| | - Vanessa Ayo-Jibunoh
- Department of Psychology, College of Staten Island, City University of New York, Staten Island, NY, USA
| | - Shelby Louis
- Department of Psychology, College of Staten Island, City University of New York, Staten Island, NY, USA
| | - Daniel Maronna
- Department of Psychology, College of Staten Island, City University of New York, Staten Island, NY, USA
| | - Rania Darwish
- Department of Psychology, College of Staten Island, City University of New York, Staten Island, NY, USA
| | - Jaquelyn Tomaio
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, New York, NY, USA
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY, USA
| | - Susana Mingote
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, New York, NY, USA
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY, USA
| | - Leora Yetnikoff
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, New York, NY, USA
- Department of Psychology, College of Staten Island, City University of New York, Staten Island, NY, USA
| |
Collapse
|
3
|
Plateau V, Baufreton J, Le Bon-Jégo M. Age-Dependent Modulation of Layer V Pyramidal Neuron Excitability in the Mouse Primary Motor Cortex by D1 Receptor Agonists and Antagonists. Neuroscience 2024; 536:21-35. [PMID: 37952579 DOI: 10.1016/j.neuroscience.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023]
Abstract
The primary motor cortex (M1) receives dopaminergic (DAergic) projections from the midbrain which play a key role in modulating motor and cognitive processes, such as motor skill learning. However, little is known at the level of individual neurons about how dopamine (DA) and its receptors modulate the intrinsic properties of the different neuronal subpopulations in M1 and if this modulation depends on age. Using immunohistochemistry, we first mapped the cells expressing the DA D1 receptor across the different layers in M1, and quantified the number of pyramidal neurons (PNs) expressing the D1 receptor in the different layers, in young and adult mice. This work reveals that the spatial distribution and the molecular profile of D1 receptor-expressing neurons (D1+) across M1 layers do not change with age. Then, combining whole-cell patch-clamp recordings and pharmacology, we explored ex vivo in young and adult mice the impact of activation or blockade of D1 receptors on D1+ PN intrinsic properties. While the bath application of the D1 receptor agonist induced an increase in the excitability of layer V PNs both in young and adult, we identified a distinct modulation of intrinsic electrical properties of layer V D1+ PNs by D1 receptor antagonist depending on the age of the animal.
Collapse
Affiliation(s)
- Valentin Plateau
- Université de Bordeaux, Institut des Maladies Neurodégénératives, 33076 Bordeaux, France; CNRS UMR 5293, Institut des Maladies Neurodégénératives, 33076 Bordeaux, France
| | - Jérôme Baufreton
- Université de Bordeaux, Institut des Maladies Neurodégénératives, 33076 Bordeaux, France; CNRS UMR 5293, Institut des Maladies Neurodégénératives, 33076 Bordeaux, France
| | - Morgane Le Bon-Jégo
- Université de Bordeaux, Institut des Maladies Neurodégénératives, 33076 Bordeaux, France; CNRS UMR 5293, Institut des Maladies Neurodégénératives, 33076 Bordeaux, France.
| |
Collapse
|
4
|
Matthiesen M, Khlaifia A, Steininger CFD, Dadabhoy M, Mumtaz U, Arruda-Carvalho M. Maturation of nucleus accumbens synaptic transmission signals a critical period for the rescue of social deficits in a mouse model of autism spectrum disorder. Mol Brain 2023; 16:46. [PMID: 37226266 DOI: 10.1186/s13041-023-01028-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/20/2023] [Indexed: 05/26/2023] Open
Abstract
Social behavior emerges early in development, a time marked by the onset of neurodevelopmental disorders featuring social deficits, including autism spectrum disorder (ASD). Although social deficits are at the core of the clinical diagnosis of ASD, very little is known about their neural correlates at the time of clinical onset. The nucleus accumbens (NAc), a brain region extensively implicated in social behavior, undergoes synaptic, cellular and molecular alterations in early life, and is particularly affected in ASD mouse models. To explore a link between the maturation of the NAc and neurodevelopmental deficits in social behavior, we compared spontaneous synaptic transmission in NAc shell medium spiny neurons (MSNs) between the highly social C57BL/6J and the idiopathic ASD mouse model BTBR T+Itpr3tf/J at postnatal day (P) 4, P6, P8, P12, P15, P21 and P30. BTBR NAc MSNs display increased spontaneous excitatory transmission during the first postnatal week, and increased inhibition across the first, second and fourth postnatal weeks, suggesting accelerated maturation of excitatory and inhibitory synaptic inputs compared to C57BL/6J mice. BTBR mice also show increased optically evoked medial prefrontal cortex-NAc paired pulse ratios at P15 and P30. These early changes in synaptic transmission are consistent with a potential critical period, which could maximize the efficacy of rescue interventions. To test this, we treated BTBR mice in either early life (P4-P8) or adulthood (P60-P64) with the mTORC1 antagonist rapamycin, a well-established intervention for ASD-like behavior. Rapamycin treatment rescued social interaction deficits in BTBR mice when injected in infancy, but did not affect social interaction in adulthood.
Collapse
Affiliation(s)
- Melina Matthiesen
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Abdessattar Khlaifia
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | | | - Maryam Dadabhoy
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Unza Mumtaz
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Maithe Arruda-Carvalho
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada.
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S3G5, Canada.
| |
Collapse
|
5
|
Peters KZ, Naneix F. The role of dopamine and endocannabinoid systems in prefrontal cortex development: Adolescence as a critical period. Front Neural Circuits 2022; 16:939235. [PMID: 36389180 PMCID: PMC9663658 DOI: 10.3389/fncir.2022.939235] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 10/14/2022] [Indexed: 01/07/2023] Open
Abstract
The prefrontal cortex plays a central role in the control of complex cognitive processes including action control and decision making. It also shows a specific pattern of delayed maturation related to unique behavioral changes during adolescence and allows the development of adult cognitive processes. The adolescent brain is extremely plastic and critically vulnerable to external insults. Related to this vulnerability, adolescence is also associated with the emergence of numerous neuropsychiatric disorders involving alterations of prefrontal functions. Within prefrontal microcircuits, the dopamine and the endocannabinoid systems have widespread effects on adolescent-specific ontogenetic processes. In this review, we highlight recent advances in our understanding of the maturation of the dopamine system and the endocannabinoid system in the prefrontal cortex during adolescence. We discuss how they interact with GABA and glutamate neurons to modulate prefrontal circuits and how they can be altered by different environmental events leading to long-term neurobiological and behavioral changes at adulthood. Finally, we aim to identify several future research directions to help highlight gaps in our current knowledge on the maturation of these microcircuits.
Collapse
Affiliation(s)
- Kate Zara Peters
- Sussex Neuroscience, School of Psychology, University of Sussex, Falmer, United Kingdom
| | - Fabien Naneix
- The Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom,*Correspondence: Fabien Naneix
| |
Collapse
|
6
|
Yang S, Tseng KY. Maturation of Corticolimbic Functional Connectivity During Sensitive Periods of Brain Development. Curr Top Behav Neurosci 2022; 53:37-53. [PMID: 34386969 DOI: 10.1007/7854_2021_239] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The maturation of key corticolimbic structures and the prefrontal cortex during sensitive periods of brain development from early life through adolescence is crucial for the acquisition of a variety of cognitive and affective processes associated with adult behavior. In this chapter, we first review how key cellular and circuit level changes during adolescence dictate the development of the prefrontal cortex and its capacity to integrate contextual and emotional information from the ventral hippocampus and the amygdala. We further discuss how afferent transmission from ventral hippocampal and amygdala inputs displays unique age-dependent trajectories that directly impact prefrontal functional maturation through adolescence. We conclude by proposing that time-sensitive strengthening of specific corticolimbic synapses is a critical contributing factor for the protracted maturation of cognitive and emotional regulation by the prefrontal cortex.
Collapse
Affiliation(s)
- Shaolin Yang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kuei Y Tseng
- Department of Anatomy and Cell Biology, University of Illinois at Chicago - College of Medicine, Chicago, IL, USA.
| |
Collapse
|
7
|
Islam KUS, Meli N, Blaess S. The Development of the Mesoprefrontal Dopaminergic System in Health and Disease. Front Neural Circuits 2021; 15:746582. [PMID: 34712123 PMCID: PMC8546303 DOI: 10.3389/fncir.2021.746582] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 09/10/2021] [Indexed: 12/18/2022] Open
Abstract
Midbrain dopaminergic neurons located in the substantia nigra and the ventral tegmental area are the main source of dopamine in the brain. They send out projections to a variety of forebrain structures, including dorsal striatum, nucleus accumbens, and prefrontal cortex (PFC), establishing the nigrostriatal, mesolimbic, and mesoprefrontal pathways, respectively. The dopaminergic input to the PFC is essential for the performance of higher cognitive functions such as working memory, attention, planning, and decision making. The gradual maturation of these cognitive skills during postnatal development correlates with the maturation of PFC local circuits, which undergo a lengthy functional remodeling process during the neonatal and adolescence stage. During this period, the mesoprefrontal dopaminergic innervation also matures: the fibers are rather sparse at prenatal stages and slowly increase in density during postnatal development to finally reach a stable pattern in early adulthood. Despite the prominent role of dopamine in the regulation of PFC function, relatively little is known about how the dopaminergic innervation is established in the PFC, whether and how it influences the maturation of local circuits and how exactly it facilitates cognitive functions in the PFC. In this review, we provide an overview of the development of the mesoprefrontal dopaminergic system in rodents and primates and discuss the role of altered dopaminergic signaling in neuropsychiatric and neurodevelopmental disorders.
Collapse
Affiliation(s)
- K Ushna S Islam
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Norisa Meli
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Bonn, Germany.,Institute of Neuropathology, Section for Translational Epilepsy Research, Medical Faculty, University of Bonn, Bonn, Germany
| | - Sandra Blaess
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
8
|
Reynolds LM, Flores C. Mesocorticolimbic Dopamine Pathways Across Adolescence: Diversity in Development. Front Neural Circuits 2021; 15:735625. [PMID: 34566584 PMCID: PMC8456011 DOI: 10.3389/fncir.2021.735625] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/17/2021] [Indexed: 12/26/2022] Open
Abstract
Mesocorticolimbic dopamine circuity undergoes a protracted maturation during adolescent life. Stable adult levels of behavioral functioning in reward, motivational, and cognitive domains are established as these pathways are refined, however, their extended developmental window also leaves them vulnerable to perturbation by environmental factors. In this review, we highlight recent advances in understanding the mechanisms underlying dopamine pathway development in the adolescent brain, and how the environment influences these processes to establish or disrupt neurocircuit diversity. We further integrate these recent studies into the larger historical framework of anatomical and neurochemical changes occurring during adolescence in the mesocorticolimbic dopamine system. While dopamine neuron heterogeneity is increasingly appreciated at molecular, physiological, and anatomical levels, we suggest that a developmental facet may play a key role in establishing vulnerability or resilience to environmental stimuli and experience in distinct dopamine circuits, shifting the balance between healthy brain development and susceptibility to psychiatric disease.
Collapse
Affiliation(s)
- Lauren M Reynolds
- Plasticité du Cerveau CNRS UMR8249, École supérieure de physique et de chimie industrielles de la Ville de Paris (ESPCI Paris), Paris, France.,Neuroscience Paris Seine CNRS UMR 8246 INSERM U1130, Institut de Biologie Paris Seine, Sorbonne Université, Paris, France
| | - Cecilia Flores
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montréal, QC, Canada
| |
Collapse
|
9
|
Klune CB, Jin B, DeNardo LA. Linking mPFC circuit maturation to the developmental regulation of emotional memory and cognitive flexibility. eLife 2021; 10:e64567. [PMID: 33949949 PMCID: PMC8099425 DOI: 10.7554/elife.64567] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 03/28/2021] [Indexed: 12/12/2022] Open
Abstract
The medial prefrontal cortex (mPFC) and its abundant connections with other brain regions play key roles in memory, cognition, decision making, social behaviors, and mood. Dysfunction in mPFC is implicated in psychiatric disorders in which these behaviors go awry. The prolonged maturation of mPFC likely enables complex behaviors to emerge, but also increases their vulnerability to disruption. Many foundational studies have characterized either mPFC synaptic or behavioral development without establishing connections between them. Here, we review this rich body of literature, aligning major events in mPFC development with the maturation of complex behaviors. We focus on emotional memory and cognitive flexibility, and highlight new work linking mPFC circuit disruption to alterations of these behaviors in disease models. We advance new hypotheses about the causal connections between mPFC synaptic development and behavioral maturation and propose research strategies to establish an integrated understanding of neural architecture and behavioral repertoires.
Collapse
Affiliation(s)
- Cassandra B Klune
- Physiology Department, David Geffen School of Medicine, UCLALos AngelesUnited States
- Neuroscience Interdepartmental Graduate Program, UCLALos AngelesUnited States
| | - Benita Jin
- Physiology Department, David Geffen School of Medicine, UCLALos AngelesUnited States
- Molecular, Cellular and Integrative Physiology Graduate Program, UCLALos AngelesUnited States
| | - Laura A DeNardo
- Physiology Department, David Geffen School of Medicine, UCLALos AngelesUnited States
| |
Collapse
|
10
|
Resolving and Rescuing Developmental Miswiring in a Mouse Model of Cognitive Impairment. Neuron 2019; 105:60-74.e7. [PMID: 31733940 PMCID: PMC6953432 DOI: 10.1016/j.neuron.2019.09.042] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 08/14/2019] [Accepted: 09/24/2019] [Indexed: 12/21/2022]
Abstract
Cognitive deficits, core features of mental illness, largely result from dysfunction of prefrontal networks. This dysfunction emerges during early development, before a detectable behavioral readout, yet the cellular elements controlling the abnormal maturation are still unknown. Here, we address this open question by combining in vivo electrophysiology, optogenetics, neuroanatomy, and behavioral assays during development in mice mimicking the dual genetic-environmental etiology of psychiatric disorders. We report that pyramidal neurons in superficial layers of the prefrontal cortex are key elements causing disorganized oscillatory entrainment of local circuits in beta-gamma frequencies. Their abnormal firing rate and timing relate to sparser dendritic arborization and lower spine density. Administration of minocycline during the first postnatal week, potentially acting via microglial cells, rescues the neuronal deficits and restores pre-juvenile cognitive abilities. Elucidation of the cellular substrate of developmental miswiring causing later cognitive deficits opens new perspectives for identification of neurobiological targets amenable to therapies. Mice mimicking the etiology of mental illness have dysregulated prefrontal network Weaker beta activation of prefrontal circuits results from superficial layers deficits Rescue of microglial function restores developing prefrontal function and behavior Early prefrontal dysfunction relates to later-emerging cognitive performance
Collapse
|
11
|
Thorpe HHA, Hamidullah S, Jenkins BW, Khokhar JY. Adolescent neurodevelopment and substance use: Receptor expression and behavioral consequences. Pharmacol Ther 2019; 206:107431. [PMID: 31706976 DOI: 10.1016/j.pharmthera.2019.107431] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2019] [Indexed: 12/18/2022]
Abstract
Adolescence is the transitional period between childhood and adulthood, during which extensive brain development occurs. Since this period also overlaps with the initiation of drug use, it is important to consider how substance use during this time might produce long-term neurobiological alterations, especially against the backdrop of developmental changes in neurotransmission. Alcohol, cannabis, nicotine, and opioids all produce marked changes in the expression and function of the neurotransmitter and receptor systems with which they interact. These acute and chronic alterations also contribute to behavioral consequences ranging from increased addiction risk to cognitive or neuropsychiatric behavioral dysfunctions. The current review provides an in-depth overview and update of the developmental changes in neurotransmission during adolescence, as well as the impact of drug exposure during this neurodevelopmental window. While most of these factors have been studied in animal models, which are the focus of this review, future longitudinal studies in humans that assess neural function and behavior will help to confirm pre-clinical findings. Furthermore, the neural changes induced by each drug should also be considered in the context of other contributing factors, such as sex. Further understanding of these consequences can help in the identification of novel approaches for preventing and reversing the neurobiological effects of adolescent substance use.
Collapse
Affiliation(s)
- Hayley H A Thorpe
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Ontario, Canada
| | - Shahnaza Hamidullah
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Ontario, Canada
| | - Bryan W Jenkins
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Ontario, Canada
| | - Jibran Y Khokhar
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Ontario, Canada.
| |
Collapse
|
12
|
Dow-Edwards D, MacMaster FP, Peterson BS, Niesink R, Andersen S, Braams BR. Experience during adolescence shapes brain development: From synapses and networks to normal and pathological behavior. Neurotoxicol Teratol 2019; 76:106834. [PMID: 31505230 DOI: 10.1016/j.ntt.2019.106834] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 07/27/2019] [Accepted: 09/06/2019] [Indexed: 12/20/2022]
Abstract
Adolescence is a period of dramatic neural reorganization creating a period of vulnerability and the possibility for the development of psychopathology. The maturation of various neural circuits during adolescence depends, to a large degree, on one's experiences both physical and psychosocial. This occurs through a process of plasticity which is the structural and functional adaptation of the nervous system in response to environmental demands, physiological changes and experiences. During adolescence, this adaptation proceeds upon a backdrop of structural and functional alterations imparted by genetic and epigenetic factors and experiences both prior to birth and during the postnatal period. Plasticity entails an altering of connections between neurons through long-term potentiation (LTP) (which alters synaptic efficiency), synaptogenesis, axonal sprouting, dendritic remodeling, neurogenesis and recruitment (Skaper et al., 2017). Although most empirical evidence for plasticity derives from studies of the sensory systems, recent studies have suggested that during adolescence, social, emotional, and cognitive experiences alter the structure and function of the networks subserving these domains of behavior. Each of these neural networks exhibits heightened vulnerability to experience-dependent plasticity during the sensitive periods which occur in different circuits and different brain regions at specific periods of development. This report will summarize some examples of adaptation which occur during adolescence and some evidence that the adolescent brain responds differently to stimuli compared to adults and children. This symposium, "Experience during adolescence shapes brain development: from synapses and networks to normal and pathological behavior" occurred during the Developmental Neurotoxicology Society/Teratology Society Annual Meeting in Clearwater Florida, June 2018. The sections will describe the maturation of the brain during adolescence as studied using imaging technologies, illustrate how plasticity shapes the structure of the brain using examples of pathological conditions such as Tourette's' syndrome and attention deficit hyperactivity disorder, and a review of the key molecular systems involved in this plasticity and how some commonly abused substances alter brain development. The role of stimulants used in the treatment of attention deficit hyperactivity disorder (ADHD) in the plasticity of the reward circuit is then described. Lastly, clinical data promoting an understanding of peer-influences on risky behavior in adolescents provides evidence for the complexity of the roles that peers play in decision making, a phenomenon different from that in the adult. Imaging studies have revealed that activation of the social network by the presence of peers at times of decision making is unique in the adolescent. Since normal brain development relies on experiences which alter the functional and structural connections between cells within circuits and networks to ultimately alter behavior, readers can be made aware of the myriad of ways normal developmental processes can be hijacked. The vulnerability of developing adolescent brain places the adolescent at risk for the development of a life time of abnormal behaviors and mental disorders.
Collapse
Affiliation(s)
- Diana Dow-Edwards
- Department of Physiology & Pharmacology, State University of New York, Downstate Medical Center, Brooklyn, NY, United States of America.
| | - Frank P MacMaster
- Departments of Psychiatry & Pediatrics, University of Calgary, Addiction and Mental Health Strategic Clinical Network, Calgary, Alberta, Canada
| | - Bradley S Peterson
- Children's Hospital Los Angeles, The Keck School of Medicine at the University of Southern California, Los Angeles, CA, United States of America
| | - Raymond Niesink
- Trimbos Institute, Netherlands Institute of Mental Health and Addiction, Utrecht, the Netherlands; Faculty of Management, Science and Technology, School of Science, Open University of the Netherlands, Heerlen, the Netherlands
| | - Susan Andersen
- McLean Hospital, Department of Psychiatry, Harvard Medical School, Boston, MA, United States of America
| | - B R Braams
- Department of Psychology, Center for Brain Science, Harvard University, Cambridge, MA, United States of America
| |
Collapse
|
13
|
Gavin WJ, Lin MH, Davies PL. Developmental trends of performance monitoring measures in 7- to 25-year-olds: Unraveling the complex nature of brain measures. Psychophysiology 2019; 56:e13365. [PMID: 30942480 PMCID: PMC6570561 DOI: 10.1111/psyp.13365] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 01/19/2019] [Accepted: 02/18/2019] [Indexed: 12/19/2022]
Abstract
This study explores how trial-to-trial latency variability contributes to the developmental trends observed in ERN amplitude found in the incorrect trials of a performance monitoring task, the visual flanker task. An Adaptive Woody filter was used to measure and correct for the trial-to-trial latency variability of the ERN in 240 participants aged 7-25 years. Using three measures of latency variability, the degree of trial-to-trial latency variability was shown to decrease as the age of the participants increased from 7 to 25 years. The success of the Adaptive Woody filter technique to remove the trial-to-trial latency variability was demonstrated in a straightforward manner by the significant changes in the measures of fit and intraindividual variability obtained before and after applying the filter. After the latency variability effects were removed and adjusted averaged ERPs were obtained, a more subtle but significant nonlinear developmental trend was still found in the amplitude of the ERN component.
Collapse
Affiliation(s)
- William J. Gavin
- Department of Molecular, Cellular & Integrative Neurosciences, Colorado State University, Fort Collins, CO, United States
| | - Mei-Heng Lin
- Department of Occupational Therapy, Colorado State University, Fort Collins, CO, United States
| | - Patricia L. Davies
- Department of Molecular, Cellular & Integrative Neurosciences, Colorado State University, Fort Collins, CO, United States
- Department of Occupational Therapy, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
14
|
Cullity ER, Madsen HB, Perry CJ, Kim JH. Postnatal developmental trajectory of dopamine receptor 1 and 2 expression in cortical and striatal brain regions. J Comp Neurol 2018; 527:1039-1055. [DOI: 10.1002/cne.24574] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/28/2018] [Accepted: 10/30/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Ellen R. Cullity
- Mental Health Theme Florey Institute of Neuroscience and Mental Health Melbourne Victoria Australia
- The Florey Department of Neuroscience and Mental Health The University of Melbourne Melbourne Australia
| | - Heather B. Madsen
- Mental Health Theme Florey Institute of Neuroscience and Mental Health Melbourne Victoria Australia
- The Florey Department of Neuroscience and Mental Health The University of Melbourne Melbourne Australia
| | - Christina J. Perry
- Mental Health Theme Florey Institute of Neuroscience and Mental Health Melbourne Victoria Australia
- The Florey Department of Neuroscience and Mental Health The University of Melbourne Melbourne Australia
| | - Jee Hyun Kim
- Mental Health Theme Florey Institute of Neuroscience and Mental Health Melbourne Victoria Australia
- The Florey Department of Neuroscience and Mental Health The University of Melbourne Melbourne Australia
| |
Collapse
|
15
|
Larson TA, O’Neill CE, Palumbo MP, Bachtell RK. Effects of adolescent caffeine consumption on cocaine self-administration and reinstatement of cocaine seeking. J Psychopharmacol 2018; 33:269881118812098. [PMID: 30484365 PMCID: PMC6766411 DOI: 10.1177/0269881118812098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Caffeine consumption by children and adolescents has risen dramatically in recent years, yet the lasting effects of caffeine consumption during adolescence remain poorly understood. AIM: These experiments explore the effects of adolescent caffeine consumption on cocaine self-administration and seeking using a rodent model. METHODS: Sprague-Dawley rats consumed caffeine for 28 days during the adolescent period. Following the caffeine consumption period, the caffeine solution was replaced with water for the remainder of the experiment. Age-matched control rats received water for the duration of the study. Behavioral testing in a cocaine self-administration procedure occurred during adulthood (postnatal days 62-82) to evaluate how adolescent caffeine exposure influenced the reinforcing properties of cocaine. Cocaine seeking was also tested during extinction training and reinstatement tests following cocaine self-administration. RESULTS: Adolescent caffeine consumption increased the acquisition of cocaine self-administration and increased performance on different schedules of reinforcement. Consumption of caffeine in adult rats did not produce similar enhancements in cocaine self-administration. Adolescent caffeine consumption also produced an upward shift in the U-shaped dose response curve on cocaine self-administration maintained on a within-session dose-response procedure. Adolescent caffeine consumption had no effect on cocaine seeking during extinction training or reinstatement of cocaine seeking by cues or cocaine. CONCLUSIONS: These findings suggest that caffeine consumption during adolescence may enhance the reinforcing properties of cocaine, leading to enhanced acquisition that may contribute to increased addiction vulnerability.
Collapse
Affiliation(s)
- Tracey A Larson
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO USA
| | - Casey E O’Neill
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO USA
| | - Michaela P Palumbo
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO USA
| | - Ryan K Bachtell
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO USA
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO USA
| |
Collapse
|
16
|
Larsen B, Luna B. Adolescence as a neurobiological critical period for the development of higher-order cognition. Neurosci Biobehav Rev 2018; 94:179-195. [PMID: 30201220 PMCID: PMC6526538 DOI: 10.1016/j.neubiorev.2018.09.005] [Citation(s) in RCA: 396] [Impact Index Per Article: 56.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/29/2018] [Accepted: 09/06/2018] [Indexed: 01/08/2023]
Abstract
The transition from adolescence to adulthood is characterized by improvements in higher-order cognitive abilities and corresponding refinements of the structure and function of the brain regions that support them. Whereas the neurobiological mechanisms that govern early development of sensory systems are well-understood, the mechanisms that drive developmental plasticity of association cortices, such as prefrontal cortex (PFC), during adolescence remain to be explained. In this review, we synthesize neurodevelopmental findings at the cellular, circuit, and systems levels in PFC and evaluate them through the lens of established critical period (CP) mechanisms that guide early sensory development. We find remarkable correspondence between these neurodevelopmental processes and the mechanisms driving CP plasticity, supporting the hypothesis that adolescent development is driven by CP mechanisms that guide the rapid development of neurobiology and cognitive ability during adolescence and their subsequent stability in adulthood. Critically, understanding adolescence as a CP not only provides a mechanism for normative adolescent development, it provides a framework for understanding the role of experience and neurobiology in the emergence of psychopathology that occurs during this developmental period.
Collapse
Affiliation(s)
- Bart Larsen
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, 15213, United States; Center for the Neural Basis of Cognition, Pittsburgh, PA, 15213, United States.
| | - Beatriz Luna
- Center for the Neural Basis of Cognition, Pittsburgh, PA, 15213, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, United States
| |
Collapse
|
17
|
Ye Y, Mastwal S, Cao VY, Ren M, Liu Q, Zhang W, Elkahloun AG, Wang KH. Dopamine is Required for Activity-Dependent Amplification of Arc mRNA in Developing Postnatal Frontal Cortex. Cereb Cortex 2018; 27:3600-3608. [PMID: 27365296 DOI: 10.1093/cercor/bhw181] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The activity-regulated gene Arc/Arg3.1 encodes a postsynaptic protein crucially involved in glutamatergic synaptic plasticity. Genetic mutations in Arc pathway and altered Arc expression in human frontal cortex have been associated with schizophrenia. Although Arc expression has been reported to vary with age, what mechanisms regulate Arc mRNA levels in frontal cortex during postnatal development remains unclear. Using quantitative mRNA analysis of mouse frontal cortical tissues, we mapped the developmental profiles of Arc expression and found that its mRNA levels are sharply amplified near the end of the second postnatal week, when mouse pups open their eyes for the first time after birth. Surprisingly, electrical stimulation of the frontal cortex before eye-opening is not sufficient to drive the amplification of Arc mRNA. Instead, this amplification needs both electrical stimulation and dopamine D1-type receptor (D1R) activation. Furthermore, visual stimuli-driven amplification of Arc mRNA is also dependent on D1R activation and dopamine neurons located in the ventral midbrain. These results indicate that dopamine is required to drive activity-dependent amplification of Arc mRNA in the developing postnatal frontal cortex and suggest that joint electrical and dopaminergic activation is essential to establish the normal expression pattern of a schizophrenia-associated gene during frontal cortical development.
Collapse
Affiliation(s)
- Yizhou Ye
- Unit on Neural Circuits and Adaptive Behaviors, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Surjeet Mastwal
- Unit on Neural Circuits and Adaptive Behaviors, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vania Yu Cao
- Unit on Neural Circuits and Adaptive Behaviors, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ming Ren
- Unit on Neural Circuits and Adaptive Behaviors, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qing Liu
- Unit on Neural Circuits and Adaptive Behaviors, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wenyu Zhang
- Unit on Neural Circuits and Adaptive Behaviors, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Abdel G Elkahloun
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kuan Hong Wang
- Unit on Neural Circuits and Adaptive Behaviors, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Lo Iacono L, Carola V. The impact of adolescent stress experiences on neurobiological development. Semin Cell Dev Biol 2018; 77:93-103. [DOI: 10.1016/j.semcdb.2017.09.040] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/18/2017] [Accepted: 09/29/2017] [Indexed: 01/23/2023]
|
19
|
Zbukvic IC, Hyun Kim J. Divergent prefrontal dopaminergic mechanisms mediate drug- and fear-associated cue extinction during adolescence versus adulthood. Eur Neuropsychopharmacol 2018; 28:1-12. [PMID: 29174948 DOI: 10.1016/j.euroneuro.2017.11.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/08/2017] [Accepted: 11/03/2017] [Indexed: 01/21/2023]
Abstract
Cue-associated learning is vital to guiding behaviour for survival. Adolescence represents a key developmental stage for perturbations in cue-related learning, including a characteristic deficit in cue extinction learning. The present review summarizes evidence from animal and human literature that cue extinction is critically mediated by prefrontal dopamine, a system that undergoes dramatic reorganization during adolescence. We propose that extinction learning and memory is governed by a developmentally dynamic balance of dopamine receptors in the prefrontal cortex, which changes across adolescence into adulthood. This is contrary to the previous idea that extinction deficits during adolescence reflect inefficiency in the same neural circuitry as adults. This leads to proposal of the novel theory that cue extinction involves divergent prefrontal dopaminergic mechanisms depending on the age of extinction.
Collapse
Affiliation(s)
- Isabel C Zbukvic
- Black Dog Institute, Randwick 2031, NSW, Australia; Behavioral Neuroscience Division, The Florey Institute of Neuroscience & Mental Health, Parkville 3051, VIC, Australia.
| | - Jee Hyun Kim
- Behavioral Neuroscience Division, The Florey Institute of Neuroscience & Mental Health, Parkville 3051, VIC, Australia; The Florey Department of Neuroscience & Mental Health, University of Melbourne, Parkville 3010, VIC, Australia
| |
Collapse
|
20
|
Zbukvic IC, Park CHJ, Ganella DE, Lawrence AJ, Kim JH. Prefrontal Dopaminergic Mechanisms of Extinction in Adolescence Compared to Adulthood in Rats. Front Behav Neurosci 2017; 11:32. [PMID: 28275342 PMCID: PMC5319962 DOI: 10.3389/fnbeh.2017.00032] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 02/10/2017] [Indexed: 01/11/2023] Open
Abstract
Adolescents with anxiety disorders attain poorer outcomes following extinction-based treatment compared to adults. Extinction deficit during adolescence has been identified to involve immaturity in the medial prefrontal cortex (mPFC). Findings from adult rodents suggest extinction involves dopamine signaling in the mPFC. This system changes dramatically during adolescence, but its role in adolescent extinction is unknown. Therefore, we investigated the role of prefrontal dopamine in extinction using Pavlovian fear conditioning in adolescent and adult rats. Using quantitative PCR (qPCR) analyses, we measured changes in dopamine receptor gene expression in the mPFC before and after extinction. We then enhanced dopamine 1 receptor (D1R) or dopamine 2 receptor (D2R) signaling in the infralimbic cortex (IL) of the mPFC using agonists at the time of extinction. Adolescent rats displayed a deficit in extinction retention compared to adults. Extinction induced a reduction in D1R compared to D2R gene expression in adolescent rats, whereas an increase of D1R compared to D2R gene expression was observed in adult rats. Acutely enhancing IL D1R signaling using SKF-81297 had no effect on extinction at either age. In contrast, acutely enhancing IL D2R signaling with quinpirole significantly enhanced long-term extinction in adolescents, and impaired within-session extinction in adults. Our results suggest a dissociated role for prefrontal dopamine in fear extinction during adolescence compared to adulthood. Findings highlight the dopamine system as a potential pharmacological target to improve extinction-based treatments for adolescents.
Collapse
Affiliation(s)
- Isabel C Zbukvic
- Developmental Psychobiology Laboratory, Behavioral Neuroscience Division, The Florey Institute of Neuroscience and Mental HealthParkville, VIC, Australia; Developmental Psychobiology Laboratory, The Florey Department of Neuroscience and Mental Health, University of MelbourneParkville, VIC, Australia
| | - Chun Hui J Park
- Developmental Psychobiology Laboratory, Behavioral Neuroscience Division, The Florey Institute of Neuroscience and Mental HealthParkville, VIC, Australia; Developmental Psychobiology Laboratory, The Florey Department of Neuroscience and Mental Health, University of MelbourneParkville, VIC, Australia
| | - Despina E Ganella
- Developmental Psychobiology Laboratory, Behavioral Neuroscience Division, The Florey Institute of Neuroscience and Mental HealthParkville, VIC, Australia; Developmental Psychobiology Laboratory, The Florey Department of Neuroscience and Mental Health, University of MelbourneParkville, VIC, Australia
| | - Andrew J Lawrence
- Developmental Psychobiology Laboratory, Behavioral Neuroscience Division, The Florey Institute of Neuroscience and Mental HealthParkville, VIC, Australia; Developmental Psychobiology Laboratory, The Florey Department of Neuroscience and Mental Health, University of MelbourneParkville, VIC, Australia
| | - Jee Hyun Kim
- Developmental Psychobiology Laboratory, Behavioral Neuroscience Division, The Florey Institute of Neuroscience and Mental HealthParkville, VIC, Australia; Developmental Psychobiology Laboratory, The Florey Department of Neuroscience and Mental Health, University of MelbourneParkville, VIC, Australia
| |
Collapse
|
21
|
Postnatal development of neurotransmitter systems and their relevance to extinction of conditioned fear. Neurobiol Learn Mem 2017; 138:252-270. [DOI: 10.1016/j.nlm.2016.10.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 10/22/2016] [Accepted: 10/31/2016] [Indexed: 12/14/2022]
|
22
|
Caballero A, Granberg R, Tseng KY. Mechanisms contributing to prefrontal cortex maturation during adolescence. Neurosci Biobehav Rev 2016; 70:4-12. [PMID: 27235076 DOI: 10.1016/j.neubiorev.2016.05.013] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/17/2016] [Accepted: 05/18/2016] [Indexed: 12/17/2022]
Abstract
Adolescence is defined as a transitional period between childhood and adulthood characterized by changes in social interaction and acquisition of mature cognitive abilities. These changes have been associated with the maturation of brain regions involved in the control of motivation, emotion, and cognition. Among these regions, the protracted development of the human prefrontal cortex during adolescence has been proposed to underlie the maturation of cognitive functions and the regulation of affective responses. Studies in animal models allow us to test the causal contribution of specific neural processes in the development of the prefrontal cortex and the acquisition of adult behavior. This review summarizes the cellular and synaptic mechanisms occurring in the rodent prefrontal cortex during adolescence as a model for understanding the changes underlying human prefrontal development.
Collapse
Affiliation(s)
- Adriana Caballero
- Department of Cellular & Molecular Pharmacology, The Chicago Medical School at Rosalind Franklin University of Medicine & Science, North Chicago, IL 60064, USA
| | - Rachel Granberg
- Department of Cellular & Molecular Pharmacology, The Chicago Medical School at Rosalind Franklin University of Medicine & Science, North Chicago, IL 60064, USA
| | - Kuei Y Tseng
- Department of Cellular & Molecular Pharmacology, The Chicago Medical School at Rosalind Franklin University of Medicine & Science, North Chicago, IL 60064, USA.
| |
Collapse
|
23
|
Norepinephrine versus dopamine and their interaction in modulating synaptic function in the prefrontal cortex. Brain Res 2016; 1641:217-33. [PMID: 26790349 DOI: 10.1016/j.brainres.2016.01.005] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 12/16/2015] [Accepted: 01/05/2016] [Indexed: 01/11/2023]
Abstract
Among the neuromodulators that regulate prefrontal cortical circuit function, the catecholamine transmitters norepinephrine (NE) and dopamine (DA) stand out as powerful players in working memory and attention. Perturbation of either NE or DA signaling is implicated in the pathogenesis of several neuropsychiatric disorders, including attention deficit hyperactivity disorder (ADHD), post-traumatic stress disorder (PTSD), schizophrenia, and drug addiction. Although the precise mechanisms employed by NE and DA to cooperatively control prefrontal functions are not fully understood, emerging research indicates that both transmitters regulate electrical and biochemical aspects of neuronal function by modulating convergent ionic and synaptic signaling in the prefrontal cortex (PFC). This review summarizes previous studies that investigated the effects of both NE and DA on excitatory and inhibitory transmissions in the prefrontal cortical circuitry. Specifically, we focus on the functional interaction between NE and DA in prefrontal cortical local circuitry, synaptic integration, signaling pathways, and receptor properties. Although it is clear that both NE and DA innervate the PFC extensively and modulate synaptic function by activating distinctly different receptor subtypes and signaling pathways, it remains unclear how these two systems coordinate their actions to optimize PFC function for appropriate behavior. Throughout this review, we provide perspectives and highlight several critical topics for future studies. This article is part of a Special Issue entitled SI: Noradrenergic System.
Collapse
|
24
|
Abstract
Adolescence is characterized by heightened risk-taking, including substance misuse. These behavioral patterns are influenced by ontogenic changes in neurotransmitter systems, particularly the dopamine system, which is fundamentally involved in the neural coding of reward and motivated approach behavior. During adolescence, this system evidences a peak in activity. At the same time, the dopamine (DA) system is neuroplastically altered by substance abuse, impacting subsequent function. Here, we describe properties of the dopamine system that change with typical adolescent development and that are altered with substance abuse. Much of this work has been gleaned from animal models due to limitations in measuring dopamine in pediatric samples. Structural and functional neuroimaging techniques have been used to examine structures that are heavily DA-innervated; they measure morphological and functional changes with age and with drug exposure. Presenting marijuana abuse as an exemplar, we consider recent findings that support an adolescent peak in DA-driven reward-seeking behavior and related deviations in motivational systems that are associated with marijuana abuse/dependence. Clinicians are advised that (1) chronic adolescent marijuana use may lead to deficiencies in incentive motivation, (2) that this state is due to marijuana's interactions with the developing DA system, and (3) that treatment strategies should be directed to remediating resultant deficiencies in goal-directed activity.
Collapse
|
25
|
Abstract
Brains systems undergo unique and specific dynamic changes at the cellular, circuit, and systems level that underlie the transition to adult-level cognitive control. We integrate literature from these different levels of analyses to propose a novel model of the brain basis of the development of cognitive control. The ability to consistently exert cognitive control improves into adulthood as the flexible integration of component processes, including inhibitory control, performance monitoring, and working memory, increases. Unique maturational changes in brain structure, supported by interactions between dopaminergic and GABAergic systems, contribute to enhanced network synchronization and an improved signal-to-noise ratio. In turn, these factors facilitate the specialization and strengthening of connectivity in networks supporting the transition to adult levels of cognitive control. This model provides a novel understanding of the adolescent period as an adaptive period of heightened experience-seeking necessary for the specialization of brain systems supporting cognitive control.
Collapse
|
26
|
Stress in adolescence and drugs of abuse in rodent models: role of dopamine, CRF, and HPA axis. Psychopharmacology (Berl) 2014; 231:1557-80. [PMID: 24370534 PMCID: PMC3969449 DOI: 10.1007/s00213-013-3369-1] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 11/18/2013] [Indexed: 01/12/2023]
Abstract
RATIONALE Research on adolescence and drug abuse increased substantially in the past decade. However, drug-addiction-related behaviors following stressful experiences during adolescence are less studied. We focus on rodent models of adolescent stress cross-sensitization to drugs of abuse. OBJECTIVES Review the ontogeny of behavior, dopamine, corticotropin-releasing factor (CRF), and the hypothalamic-pituitary-adrenal (HPA) axis in adolescent rodents. We evaluate evidence that stressful experiences during adolescence engender hypersensitivity to drugs of abuse and offer potential neural mechanisms. RESULTS AND CONCLUSIONS Much evidence suggests that final maturation of behavior, dopamine systems, and HPA axis occurs during adolescence. Stress during adolescence increases amphetamine- and ethanol-stimulated locomotion, preference, and self-administration under many conditions. The influence of adolescent stress on subsequent cocaine- and nicotine-stimulated locomotion and preference is less clear. The type of adolescent stress, temporal interval between stress and testing, species, sex, and the drug tested are key methodological determinants for successful cross-sensitization procedures. The sensitization of the mesolimbic dopamine system is proposed to underlie stress cross-sensitization to drugs of abuse in both adolescents and adults through modulation by CRF. Reduced levels of mesocortical dopamine appear to be a unique consequence of social stress during adolescence. Adolescent stress may reduce the final maturation of cortical dopamine through D2 dopamine receptor regulation of dopamine synthesis or glucocorticoid-facilitated pruning of cortical dopamine fibers. Certain rodent models of adolescent adversity are useful for determining neural mechanisms underlying the cross-sensitization to drugs of abuse.
Collapse
|
27
|
Stanis JJ, Andersen SL. Reducing substance use during adolescence: a translational framework for prevention. Psychopharmacology (Berl) 2014; 231:1437-53. [PMID: 24464527 PMCID: PMC3969413 DOI: 10.1007/s00213-013-3393-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 11/30/2013] [Indexed: 11/30/2022]
Abstract
RATIONALE Most substance use is initiated during adolescence when substantial development of relevant brain circuitry is still rapidly maturing. Developmental differences in reward processing, behavioral flexibility, and self-regulation lead to changes in resilience or vulnerability to drugs of abuse depending on exposure to risk factors. Intervention and prevention approaches to reducing addiction in teens may be able to capitalize on malleable brain systems in a predictable manner. OBJECTIVE This review will highlight what is known about how factors that increase vulnerability to addiction, including developmental stage, exposure to early life adversity (ranging from abuse, neglect, and bullying), drug exposure, and genetic predisposition, impact the development of relevant systems. RESULTS AND CONCLUSIONS Appropriate, early intervention may restore the normal course of an abnormal trajectory and reduce the likelihood of developing a substance use disorder (SUD) later in life. A considerable amount is known about the functional neuroanatomy and/or pharmacology of risky behaviors based on clinical and preclinical studies, but relatively little has been directly translated to reduce their impact on addiction in high-risk children or teenagers. An opportunity exists to effectively intervene before adolescence when substance use is likely to emerge.
Collapse
Affiliation(s)
- Jessica J Stanis
- Laboratory of Developmental Neuropharmacology, McLean Hospital and Department of Psychiatry, Harvard Medical School, Mailstop 333, 115 Mill Street, Belmont, MA, 02478, USA
| | | |
Collapse
|
28
|
Yetnikoff L, Reichard RA, Schwartz ZM, Parsely KP, Zahm DS. Protracted maturation of forebrain afferent connections of the ventral tegmental area in the rat. J Comp Neurol 2014; 522:1031-47. [PMID: 23983069 PMCID: PMC4217282 DOI: 10.1002/cne.23459] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 07/30/2013] [Accepted: 08/14/2013] [Indexed: 01/21/2023]
Abstract
The mesocorticolimbic dopamine system has long attracted the interest of researchers concerned with the unique gamut of behavioral and mental health vulnerabilities associated with adolescence. Accordingly, the development of the mesocorticolimbic system has been studied extensively, but almost exclusively with regard to dopaminergic output, particularly in the nucleus accumbens and medial prefrontal cortex. To the contrary, the ontogeny of inputs to the ventral tegmental area (VTA), the source of mesocorticolimbic dopamine, has been neglected. This is not a trivial oversight, as the activity of VTA neurons, which reflects their capacity to transmit information about salient events, is sensitively modulated by inputs. Here, we assessed the development of VTA afferent connections using the β subunit of cholera toxin (Ctβ) as a retrograde axonal tracer in adolescent (postnatal day 39) and early adult (8-9-week-old) rats. After intra-VTA injections of Ctβ, adolescent and early adult animals exhibited qualitatively similar distributions of retrogradely labeled neurons in the sense that VTA-projecting neurons were present at all of the same rostrocaudal levels in all of the same structures in both age groups. However, quantitation of retrogradely labeled neurons revealed that adolescent brains, compared with early adult brains, had significantly fewer VTA-projecting neurons preferentially within an interconnected network of cortical and striatopallidal forebrain structures. These findings provide a novel perspective on the development of the mesocorticolimbic dopamine system and may have important implications for age-dependent specificity in the function of this system, particularly with regard to adolescent impulsivity and mental health vulnerabilities.
Collapse
Affiliation(s)
- Leora Yetnikoff
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104
| | - Rhett A. Reichard
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104
| | - Zachary M. Schwartz
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104
| | - Kenneth P. Parsely
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104
| | - Daniel S. Zahm
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104
| |
Collapse
|
29
|
Holder MK, Blaustein JD. Puberty and adolescence as a time of vulnerability to stressors that alter neurobehavioral processes. Front Neuroendocrinol 2014; 35:89-110. [PMID: 24184692 PMCID: PMC3946873 DOI: 10.1016/j.yfrne.2013.10.004] [Citation(s) in RCA: 167] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 10/03/2013] [Accepted: 10/23/2013] [Indexed: 01/30/2023]
Abstract
Puberty and adolescence are major life transitions during which an individual's physiology and behavior changes from that of a juvenile to that of an adult. Here we review studies documenting the effects of stressors during pubertal and adolescent development on the adult brain and behavior. The experience of complex or compound stressors during puberty/adolescence generally increases stress reactivity, increases anxiety and depression, and decreases cognitive performance in adulthood. These behavioral changes correlate with decreased hippocampal volumes and alterations in neural plasticity. Moreover, stressful experiences during puberty disrupt behavioral responses to gonadal hormones both in sexual performance and on cognition and emotionality. These behavioral changes correlate with altered estrogen receptor densities in some estrogen-concentrating brain areas, suggesting a remodeling of the brain's response to hormones. A hypothesis is presented that activation of the immune system results in chronic neuroinflammation that may mediate the alterations of hormone-modulated behaviors in adulthood.
Collapse
Affiliation(s)
- Mary K Holder
- Neuroscience and Behavior Program, Tobin Hall, University of Massachusetts, Amherst, MA 01003-9271, USA; Center for Neuroendocrine Studies, Tobin Hall, University of Massachusetts, Amherst, MA 01003-9271, USA.
| | - Jeffrey D Blaustein
- Neuroscience and Behavior Program, Tobin Hall, University of Massachusetts, Amherst, MA 01003-9271, USA; Center for Neuroendocrine Studies, Tobin Hall, University of Massachusetts, Amherst, MA 01003-9271, USA.
| |
Collapse
|
30
|
Markham JA, Mullins SE, Koenig JI. Periadolescent maturation of the prefrontal cortex is sex-specific and is disrupted by prenatal stress. J Comp Neurol 2013; 521:1828-43. [PMID: 23172080 DOI: 10.1002/cne.23262] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Revised: 09/01/2012] [Accepted: 11/06/2012] [Indexed: 12/19/2022]
Abstract
The prefrontal cortex (PFC) undergoes dramatic, sex-specific maturation during adolescence. Adolescence is a vulnerable window for developing mental illnesses that show significant sexual dimorphisms. Gestational stress is associated with increased risk for both schizophrenia, which is more common among men, and cognitive deficits. We have shown that male, but not female, rats exposed to prenatal stress develop postpubertal deficits in cognitive behaviors supported by the prefrontal cortex. Here we tested the hypothesis that repeated variable prenatal stress during the third week of rat gestation disrupts periadolescent development of prefrontal neurons in a sex-specific fashion. Using Golgi-Cox stained tissue, we compared dendritic arborization and spine density of prelimbic layer III neurons in prenatally stressed and control animals at juvenile (day 20), prepubertal (day 30), postpubertal (day 56), and adult (day 90) ages (N = 115). Dendritic ramification followed a sex-specific pattern that was disrupted during adolescence in prenatally stressed males, but not in females. In contrast, the impact of prenatal stress on the female PFC was not evident until adulthood. Prenatal stress also caused reductions in brain and body weights, and the latter effect was more pronounced among males. Additionally, there was a trend toward reduced testosterone levels for adult prenatally stressed males. Our findings indicate that, similarly to humans, the rat PFC undergoes sex-specific development during adolescence and furthermore that this process is disrupted by prenatal stress. These findings may be relevant to both the development of normal sex differences in cognition as well as differential male-female vulnerability to psychiatric conditions.
Collapse
Affiliation(s)
- Julie A Markham
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland-Baltimore School of Medicine, Baltimore, Maryland 21228, USA.
| | | | | |
Collapse
|
31
|
Yi F, Zhang XH, Yang CR, Li BM. Contribution of dopamine d1/5 receptor modulation of post-spike/burst afterhyperpolarization to enhance neuronal excitability of layer v pyramidal neurons in prepubertal rat prefrontal cortex. PLoS One 2013; 8:e71880. [PMID: 23977170 PMCID: PMC3748086 DOI: 10.1371/journal.pone.0071880] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Accepted: 07/09/2013] [Indexed: 12/02/2022] Open
Abstract
Dopamine (DA) receptors in the prefrontal cortex (PFC) modulate both synaptic and intrinsic plasticity that may contribute to cognitive processing. However, the ionic basis underlying DA actions to enhance neuronal plasticity in PFC remains ill-defined. Using whole-cell patch-clamp recordings in layer V-VI pyramidal cells in prepubertal rat PFC, we showed that DA, via activation of D1/5, but not D2/3/4, receptors suppress a Ca(2+)-dependent, apamin-sensitive K(+) channel that mediates post-spike/burst afterhyperpolarization (AHP) to enhance neuronal excitability of PFC neurons. This inhibition is not dependent on HCN channels. The D1/5 receptor activation also enhanced an afterdepolarizing potential (ADP) that follows the AHP. Additional single-spike analyses revealed that DA or D1/5 receptor activation suppressed the apamin-sensitive post-spike mAHP, further contributing to the increase in evoked spike firing to enhance the neuronal excitability. Taken together, the D1/5 receptor modulates intrinsic mechanisms that amplify a long depolarizing input to sustain spike firing outputs in pyramidal PFC neurons.
Collapse
Affiliation(s)
- Feng Yi
- Institute of Neurobiology and State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xue-Han Zhang
- Institute of Neurobiology and State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Charles R. Yang
- CNS Pharmacology and Ion Channel, Shanghai Chempartner Co. Ltd., Shanghai, China
| | - Bao-ming Li
- Institute of Neurobiology and State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
- Center for Neuropsychiatric Diseases, Institute of Life Science, Nanchang University, Nanchang, China
| |
Collapse
|
32
|
Hofford RS, Wellman PJ, Eitan S. Morphine alters the locomotor responses to a D2/D3 dopamine receptor agonist differentially in adolescent and adult mice. J Psychopharmacol 2012; 26:1355-65. [PMID: 22522973 DOI: 10.1177/0269881112443741] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The D2-like dopamine receptors mediate the emotional/aversive state during morphine withdrawal. Given age-dependent differences in the affective responses to withdrawal, this study examined whether the response to dopamine receptor agonists is altered differentially across ages following morphine administration. Adolescent and adult mice were injected with morphine (twice daily, 10-40 mg/kg, s.c.) or saline for 6 days. Subsequently, they were examined for their locomotor response to quinpirole, a D2/D3 receptor agonist, and SKF 38393, a D1 receptor agonist. Quinpirole dose-dependently reduced locomotion in drug-naïve animals. Initial suppression was also observed in morphine-treated animals, but was followed by enhanced locomotion. Notably, this enhanced locomotion was markedly greater in adolescents than adults. Quinpirole-induced hypo-locomotion is thought to be mediated by the presynaptic D2Short receptors, whereas its activating effect is mediated by postsynaptic D2Long/D3 receptors. This suggests that following morphine administration, the postsynaptic, but not the presynaptic, dopaminergic signaling is differentially modulated across ages. This locomotor supersensitivity was not observed for SKF 38393, a D1 dopamine receptor agonist. The D2/D3 receptors are involved in the pathophysiology of many mental illnesses. Thus, this study offers a potential explanation for the increased psychiatric disorder co-morbidities when drug use begins during adolescence.
Collapse
Affiliation(s)
- Rebecca S Hofford
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX, USA
| | | | | |
Collapse
|
33
|
Coppens CM, Siripornmongcolchai T, Wibrand K, Alme MN, Buwalda B, de Boer SF, Koolhaas JM, Bramham CR. Social Defeat during Adolescence and Adulthood Differentially Induce BDNF-Regulated Immediate Early Genes. Front Behav Neurosci 2011; 5:72. [PMID: 22065953 PMCID: PMC3206404 DOI: 10.3389/fnbeh.2011.00072] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 10/11/2011] [Indexed: 01/19/2023] Open
Abstract
Stressful life events generally enhance the vulnerability for the development of human psychopathologies such as anxiety disorders and depression. The incidence rates of adult mental disorders steeply rises during adolescence in parallel with a structural and functional reorganization of the neural circuitry underlying stress reactivity. However, the mechanisms underlying susceptibility to stress and manifestation of mental disorders during adolescence are little understood. We hypothesized that heightened sensitivity to stress during adolescence reflects age-dependent differences in the expression of activity-dependent genes involved in synaptic plasticity. Therefore, we compared the effect of social stress during adolescence with social stress in adulthood on the expression of a panel of genes linked to induction of long-term potentiation (LTP) and brain-derived neurotrophic factor (BDNF) signaling. We show that social defeat during adolescence and adulthood differentially regulates expression of the immediate early genes BDNF, Arc, Carp, and Tieg1, as measured by qPCR in tissue lysates from prefrontal cortex, nucleus accumbens, and hippocampus. In the hippocampus, mRNA levels for all four genes were robustly elevated following social defeat in adolescence, whereas none were induced by defeat in adulthood. The relationship to coping style was also examined using adult reactive and proactive coping rats. Gene expression levels of reactive and proactive animals were similar in the prefrontal cortex and hippocampus. However, a trend toward a differential expression of BDNF and Arc mRNA in the nucleus accumbens was detected. BDNF mRNA was increased in the nucleus accumbens of proactive defeated animals, whereas the expression level in reactive defeated animals was comparable to control animals. The results demonstrate striking differences in immediate early gene expression in response to social defeat in adolescent and adult rats.
Collapse
Affiliation(s)
- Caroline M Coppens
- Department of Behavioural Physiology, University of Groningen Groningen, Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Trainor BC. Stress responses and the mesolimbic dopamine system: social contexts and sex differences. Horm Behav 2011; 60:457-69. [PMID: 21907202 PMCID: PMC3217312 DOI: 10.1016/j.yhbeh.2011.08.013] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 08/19/2011] [Accepted: 08/20/2011] [Indexed: 10/17/2022]
Abstract
Organisms react to threats with a variety of behavioral, hormonal, and neurobiological responses. The study of biological responses to stress has historically focused on the hypothalamic-pituitary-adrenal axis, but other systems such as the mesolimbic dopamine system are involved. Behavioral neuroendocrinologists have long recognized the importance of the mesolimbic dopamine system in mediating the effects of hormones on species specific behavior, especially aspects of reproductive behavior. There has been less focus on the role of this system in the context of stress, perhaps due to extensive data outlining its importance in reward or approach-based contexts. However, there is steadily growing evidence that the mesolimbic dopamine neurons have critical effects on behavioral responses to stress. Most of these data have been collected from experiments using a small number of animal model species under a limited set of contexts. This approach has led to important discoveries, but evidence is accumulating that mesolimbic dopamine responses are context dependent. Thus, focusing on a limited number of species under a narrow set of controlled conditions constrains our understanding of how the mesolimbic dopamine system regulates behavior in response to stress. Both affiliative and antagonistic social interactions have important effects on mesolimbic dopamine function, and there is preliminary evidence for sex differences as well. This review will highlight the benefits of expanding this approach, and focus on how social contexts and sex differences can impact mesolimbic dopamine stress responses.
Collapse
Affiliation(s)
- Brian C Trainor
- Department of Psychology, University of California, 1 Shields Ave., Davis, CA 95616, USA
| |
Collapse
|
35
|
Abolition of the behavioral phenotype of adult netrin-1 receptor deficient mice by exposure to amphetamine during the juvenile period. Psychopharmacology (Berl) 2011; 217:505-14. [PMID: 21523346 DOI: 10.1007/s00213-011-2312-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Accepted: 04/07/2011] [Indexed: 10/18/2022]
Abstract
RATIONALE Netrin-1 guidance cues contribute to amphetamine-induced plasticity of the adult mesocorticolimbic dopamine system in rodents. The netrin-1 receptor, deleted in colorectal cancer (DCC), is upregulated by repeated amphetamine treatment selectively in the ventral tegmental area (VTA) of adult rats and wild-type mice. Furthermore, adult dcc heterozygous mice fail to show amphetamine-induced increases in VTA DCC expression and do not develop sensitization to this drug. OBJECTIVES The effects of netrin-1 receptor signaling on mesocorticolimbic dopamine system function change across development. However, the effects of AMPH on DCC receptor regulation and behavioral sensitization before puberty have not been determined. Here we examined whether (1) repeated amphetamine treatment would also alter DCC expression in juvenile rats and wild-type mice, and (2) dcc heterozygotes treated with amphetamine during the juvenile period (PND 22-32) would develop behavioral sensitization to this drug. RESULTS Repeated amphetamine downregulates DCC expression selectively in the VTA of juvenile rodents. Moreover, the behavioral phenotype of adult dcc heterozygous mice is not present before puberty and is abolished by amphetamine treatment during the juvenile period. Remarkably, adult dcc heterozygotes pretreated with amphetamine as juveniles no longer exhibit reduced DCC expression in the VTA compared to wild-type controls. CONCLUSIONS Our results indicate that netrin-1 receptor signaling may be a key factor in determining individual differences in vulnerability to the behaviorally sensitizing effects of amphetamine at different ages. Moreover, they suggest that the juvenile period marks a window of vulnerability during which exposure to stimulant drugs can reverse the behavioral phenotype of adult dcc heterozygous mice.
Collapse
|
36
|
Brenhouse HC, Andersen SL. Developmental trajectories during adolescence in males and females: a cross-species understanding of underlying brain changes. Neurosci Biobehav Rev 2011; 35:1687-703. [PMID: 21600919 PMCID: PMC3134153 DOI: 10.1016/j.neubiorev.2011.04.013] [Citation(s) in RCA: 262] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Revised: 04/14/2011] [Accepted: 04/21/2011] [Indexed: 11/20/2022]
Abstract
Adolescence is a transitional period between childhood and adulthood that encompasses vast changes within brain systems that parallel some, but not all, behavioral changes. Elevations in emotional reactivity and reward processing follow an inverted U shape in terms of onset and remission, with the peak occurring during adolescence. However, cognitive processing follows a more linear course of development. This review will focus on changes within key structures and will highlight the relationships between brain changes and behavior, with evidence spanning from functional magnetic resonance imaging (fMRI) in humans to molecular studies of receptor and signaling factors in animals. Adolescent changes in neuronal substrates will be used to understand how typical and atypical behaviors arise during adolescence. We draw upon clinical and preclinical studies to provide a neural framework for defining adolescence and its role in the transition to adulthood.
Collapse
Affiliation(s)
- Heather C. Brenhouse
- Laboratory of Developmental Neuropharmacology McLean Hospital and Department of Psychiatry, Harvard Medical School
| | - Susan L. Andersen
- Laboratory of Developmental Neuropharmacology McLean Hospital and Department of Psychiatry, Harvard Medical School
| |
Collapse
|
37
|
Privitera GJ, Zavala AR, Sanabria F, Sotak KL. High fat diet intake during pre and periadolescence impairs learning of a conditioned place preference in adulthood. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2011; 7:21. [PMID: 21703027 PMCID: PMC3146828 DOI: 10.1186/1744-9081-7-21] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 06/26/2011] [Indexed: 12/02/2022]
Abstract
BACKGROUND Brain regions that mediate learning of a conditioned place preference (CPP) undergo significant development in pre and periadolescence. Consuming a high fat (HF) diet during this developmental period and into adulthood can lead to learning impairments in rodents. The present study tested whether HF diet intake, consumed only in pre and periadolescence, would be sufficient to cause impairments using a CPP procedure. METHODS Rats were randomly assigned to consume a HF or a low fat (LF) diet during postnatal days (PD) 21-40 and were then placed back on a standard lab chow diet. A 20-day CPP procedure, using HF Cheetos® as the unconditioned stimulus (US), began either the next day (PD 41) or 40 days later (PD 81). A separate group of adult rats were given the HF diet for 20 days beginning on PD 61, and then immediately underwent the 20-day CPP procedure beginning on PD 81. RESULTS Pre and periadolescent exposure to a LF diet or adult exposure to a HF diet did not interfere with the development of a HF food-induced CPP, as these groups exhibited robust preferences for the HF Cheetos® food-paired compartment. However, pre and periadolescent exposure to the HF diet impaired the development of a HF food-induced CPP regardless of whether it was assessed immediately or 40 days after the exposure to the HF diet, and despite showing increased consumption of the HF Cheetos® in conditioning. CONCLUSIONS Intake of a HF diet, consumed only in pre and periadolescence, has long-lasting effects on learning that persist into adulthood.
Collapse
Affiliation(s)
- Gregory J Privitera
- Saint Bonaventure University, Department of Psychology, 3261 West State Street, Saint Bonaventure, NY 14778, USA
| | - Arturo R Zavala
- California State University, Long Beach, Department of Psychology, 1250 Bellflower Blvd., Long Beach, CA 90840, USA
| | - Federico Sanabria
- Arizona State University, Department of Psychology, P.O. Box 871104, Tempe, AZ 85287, USA
| | - Kristin L Sotak
- Saint Bonaventure University, Department of Psychology, 3261 West State Street, Saint Bonaventure, NY 14778, USA
| |
Collapse
|
38
|
Levant B, Zarcone TJ, Davis PF, Ozias MK, Fowler SC. Differences in methylphenidate dose response between periadolescent and adult rats in the familiar arena-novel alcove task. J Pharmacol Exp Ther 2011; 337:83-91. [PMID: 21205916 PMCID: PMC3063734 DOI: 10.1124/jpet.110.174425] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 12/10/2010] [Indexed: 11/22/2022] Open
Abstract
Methylphenidate is a psychostimulant widely used in the treatment of attention deficit hyperactivity disorder. In this study, the effects of two nonstereotypy-inducing doses of methylphenidate (2.5 and 5.0 mg/kg s.c.) were examined in periadolescent [postnatal days (P) 35 and 42] and young adult (P70), male Long-Evans rats using a three-period locomotor activity paradigm that affords inferences about exploration, habituation, and attention to a novel stimulus (an "alcove") in a familiar environment in a single test session. In the first test period, P35 and P42 rats were more active than P70 rats, and methylphenidate increased locomotion in a dose-related manner. The introduction of a novel spatial stimulus in the third test period revealed a significant interaction of dose and age such that P70 rats exhibited dose-related increases in distance traveled, but P35 rats did not. Furthermore, methylphenidate dose-relatedly disrupted the rats' tendency to spend increasing amounts of time in the alcove across the test period at P70 but not at P35. Brain and serum methylphenidate concentrations were significantly lower at P35 than at P70, with intermediate levels at P42. Developmental differences in dopaminergic neurochemistry were also observed, including increased dopamine content in the caudate-putamen, nucleus accumbens, and frontal cortex and decreased densities of D(1)-like receptors in the frontal cortex in P70 than in P42 rats. These results raise the possibility that children and adults may respond differently when treated with this drug, particularly in situations involving response to novelty and that these effects involve developmental differences in pharmacokinetics and dopaminergic neurochemistry.
Collapse
Affiliation(s)
- Beth Levant
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Mail Stop 1018, 3901 Rainbow Blvd., Kansas City, KS 66160, USA.
| | | | | | | | | |
Collapse
|
39
|
Heng LJ, Markham JA, Hu XT, Tseng KY. Concurrent upregulation of postsynaptic L-type Ca(2+) channel function and protein kinase A signaling is required for the periadolescent facilitation of Ca(2+) plateau potentials and dopamine D1 receptor modulation in the prefrontal cortex. Neuropharmacology 2011; 60:953-62. [PMID: 21288471 DOI: 10.1016/j.neuropharm.2011.01.041] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Revised: 01/17/2011] [Accepted: 01/24/2011] [Indexed: 01/09/2023]
Abstract
Further understanding of how prefrontal cortex (PFC) circuit change during postnatal development is of great interest due to its role in working memory and decision-making, two cognitive abilities that are refined late in adolescence and become altered in schizophrenia. While it is evident that dopamine facilitation of glutamate responses occurs during adolescence in the PFC, little is known about the cellular mechanisms that support these changes. Among them, a developmental facilitation of postsynaptic Ca(2+) function is of particular interest given its role in coordinating neuronal ensembles, a process thought to contribute to maturation of PFC function. Here we conducted whole-cell patch clamp recordings of deep-layer pyramidal neurons in PFC brain slices and determined how somatic-evoked Ca(2+)-mediated plateau depolarizations change throughout postnatal day (PD) 25 (juvenile) to adulthood (PD 80). Postsynaptic Ca(2+) potentials in the PFC increase in duration throughout postnatal development. A remarkable shift from short to prolonged depolarizations was observed after PD 40. This change is reflected by an enhancement of L-type Ca(2+) channel function and postsynaptic PKA signaling. We speculate that such a protracted developmental facilitation of Ca(2+) response in the PFC may contribute to improvement of working memory performance through adolescence.
Collapse
Affiliation(s)
- Li-Jun Heng
- Department of Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Rd., North Chicago, IL 60064, USA
| | | | | | | |
Collapse
|
40
|
Sui ZY, Li CR, Huang GB, Kwon YB, Kim KW, Lee KH, Chung YC. Age-specific effects of 6-hydroxydopamine lesions of the rat medial prefrontal cortex on stress-induced c-fos expression in subcortical areas. Prog Neuropsychopharmacol Biol Psychiatry 2010; 34:1054-9. [PMID: 20677375 DOI: 10.1016/j.pnpbp.2010.05.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
As adolescence is a critical period when dopaminergic neuronal maturation peaks, we hypothesized that 6-hydroxydopamine (OHDA) lesions of the medial prefrontal cortex (mPFC) in adolescent rats would have more negative effects than lesions in adult rats. Therefore, we investigated the effects of 6-OHDA lesions of the mPFC in adolescent and adult rats on stress-induced c-fos expression in the brain. Adolescent and adult Sprague-Dawley rats, aged 4 and 7 weeks on arrival, respectively, were studied. 6-OHDA (8.0 microg) for the lesion groups and ascorbic acid for the sham groups were injected bilaterally into the mPFC. All animals were pretreated with desipramine 30 min before being anesthetized. The control group did not undergo any surgery-related procedure except the desipramine injection. After recovery for 1 week, the rats were subjected to restraint stress for 1 h. Immediately after the stress, the rats were killed and c-fos immunohistochemistry was examined. The c-fos expression in the nucleus accumbens core (AcbC), nucleus accumbens shell (AcbSh), CA1, CA3, dentate gyrus (DG), central amygdaloid (Ce), basolateral amygdaloid (BL), and temporal cortex (Tc) was compared. Adolescent rats with 6-OHDA lesions subjected to restraint stress had greater c-fos expression in the AcbC, AcbSh, DG, Ce, BL, and Tc, compared to the sham and control groups, whereas these differences were not observed among the adult groups. These results suggest that a hypodopaminergic state in the mPFC of adolescent rats, but not adult rats, is related to increased sensitivity to stress, suggesting that damage to or maldevelopment of dopaminergic neurons during adolescence has an age-specific effect. Further research is warranted to investigate the mechanism of the age-specific effect of 6-OHDA lesions of the mPFC.
Collapse
Affiliation(s)
- Zhi Yan Sui
- Department of Psychiatry, Chonbuk National University Medical School and Institute for Medical Sciences, San 2-20 Geumam-dong, Deokjin-gu, Jeonju 561-756, South Korea
| | | | | | | | | | | | | |
Collapse
|
41
|
Li CR, Huang GB, Sui ZY, Han EH, Chung YC. Effects of 6-hydroxydopamine lesioning of the medial prefrontal cortex on social interactions in adolescent and adult rats. Brain Res 2010; 1346:183-9. [PMID: 20513371 DOI: 10.1016/j.brainres.2010.05.064] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Revised: 05/20/2010] [Accepted: 05/20/2010] [Indexed: 10/19/2022]
Abstract
Bilateral depletion of dopamine (DA) in the medial prefrontal cortex (mPFC) following local infusions of 6-hydroxydopamine (6-OHDA) was reported to affect mesolimbic DA neurotransmission and augment spontaneous and amphetamine-induced locomotion. However, the effects of 6-OHDA lesioning of the mPFC of adolescent rats have never been investigated. Given that dopaminergic neurons reach the peak of maturation during adolescence, we hypothesized that 6-OHDA lesioning of the mPFC during adolescence would have greater impact on subsequent behavioral parameters than would such lesioning during adulthood. The aim of this study was to investigate the effects of 6-OHDA lesioning of the mPFC on the open-field activities and novel investigative and socially interactive behaviors of adolescent and adult rats. Using a stereotaxic apparatus, 6-OHDA (8.0 microg) was injected bilaterally into the mPFC of adolescent and adult rats. After a 1-week recovery period, rats were placed in an open-field chamber, and spontaneous locomotion and other behaviors were monitored. Next, a novel toy was place in the center and behavioral responses were observed. One day later, socially interactive behaviors were measured by placing the lesioned rats into a cage with four unfamiliar rats matched for age. The tests of locomotor activity and novel investigative behaviors revealed no significant differences between the lesioned and sham groups of adolescent or adult rats. Grooming and socially interactive behaviors were significantly lower in the adolescent and adult lesioned groups than in each sham group. Interestingly, we observed more extensive impairment in socially interactive behaviors among the adolescent lesioned rats compared to the adult lesioned rats. The present study indicates that DA depletion in the mPFC causes significantly reduced grooming and socially interactive behaviors; this phenomenon may be comparable to the negative symptoms observed in schizophrenia. Further research is warranted to investigate the mechanisms underpinning the detrimental effects of 6-OHDA lesioning of the mPFC on social behaviors.
Collapse
Affiliation(s)
- Chun-Rong Li
- Department of Psychiatry, Chonbuk National University Medical School and Institute for Medical Sciences, Jeonju, South Korea
| | | | | | | | | |
Collapse
|
42
|
Wahlstrom D, White T, Luciana M. Neurobehavioral evidence for changes in dopamine system activity during adolescence. Neurosci Biobehav Rev 2009; 34:631-48. [PMID: 20026110 DOI: 10.1016/j.neubiorev.2009.12.007] [Citation(s) in RCA: 166] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Revised: 12/10/2009] [Accepted: 12/12/2009] [Indexed: 11/26/2022]
Abstract
Human adolescence has been characterized by increases in risk-taking, emotional lability, and deficient patterns of behavioral regulation. These behaviors have often been attributed to changes in brain structure that occur during this developmental period, notably alterations in gray and white matter that impact synaptic architecture in frontal, limbic, and striatal regions. In this review, we provide a rationale for considering that these behaviors may be due to changes in dopamine system activity, particularly overactivity, during adolescence relative to either childhood or adulthood. This rationale relies on animal data due to limitations in assessing neurochemical activity more directly in juveniles. Accordingly, we also present a strategy that incorporates molecular genetic techniques to infer the status of the underlying tone of the dopamine system across developmental groups. Implications for the understanding of adolescent behavioral development are discussed.
Collapse
Affiliation(s)
- Dustin Wahlstrom
- Department of Psychology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
43
|
Wahlstrom D, Collins P, White T, Luciana M. Developmental changes in dopamine neurotransmission in adolescence: behavioral implications and issues in assessment. Brain Cogn 2009; 72:146-59. [PMID: 19944514 DOI: 10.1016/j.bandc.2009.10.013] [Citation(s) in RCA: 211] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Accepted: 10/22/2009] [Indexed: 12/28/2022]
Abstract
Adolescence is characterized by increased risk-taking, novelty-seeking, and locomotor activity, all of which suggest a heightened appetitive drive. The neurotransmitter dopamine is typically associated with behavioral activation and heightened forms of appetitive behavior in mammalian species, and this pattern of activation has been described in terms of a neurobehavioral system that underlies incentive-motivated behavior. Adolescence may be a time of elevated activity within this system. This review provides a summary of changes within cortical and subcortical dopaminergic systems that may account for changes in cognition and affect that characterize adolescent behavior. Because there is a dearth of information regarding neurochemical changes in human adolescents, models for assessing links between neurochemical activity and behavior in human adolescents will be described using molecular genetic techniques. Furthermore, we will suggest how these techniques can be combined with other methods such as pharmacology to measure the impact of dopamine activity on behavior and how this relation changes through the lifespan.
Collapse
Affiliation(s)
- Dustin Wahlstrom
- Department of Psychology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
44
|
Grant A, Speed Z, Labelle-Dumais C, Flores C. Post-pubertal emergence of a dopamine phenotype in netrin-1 receptor-deficient mice. Eur J Neurosci 2009; 30:1318-28. [PMID: 19788579 DOI: 10.1111/j.1460-9568.2009.06919.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
During the pubertal period the mesocortical dopamine (DA) system undergoes substantial reorganization of neuronal connectivity and functional refinement. Netrins are guidance cues involved in the organization of neuronal circuitry. We have previously shown that adult mice that develop with reduced levels of the netrin-1 receptor [deleted in colorectal cancer (DCC)] display selective reorganization of mesocortical DA circuitry, show enhanced mesocortical DA function and exhibit a behavioural phenotype opposite to that observed in animal models of schizophrenia. Here we assess whether the dcc behavioural and DA phenotypes are present prior to the maturation of the mesocortical DA system by comparing dcc-heterozygous and wild-type mice at the post-weaning and peri-pubertal periods on various indices of DA function. At both the post-weaning and peri-pubertal ages, but unlike in adulthood, dcc-heterozygous and wild-type mice show no differences in the number of midbrain DA neurones or in tyrosine hydroxylase protein levels in the medial prefrontal cortex. Furthermore, the elevated baseline concentration of mesocortical DA and DA metabolites observed in adult dcc-heterozygous mice is not present in either post-weanling or peri-pubertal mice. Interestingly, post-weanling, but not peri-pubertal, dcc-heterozygous mice show greater baseline concentrations of DA metabolites in the nucleus accumbens, opposite to what was observed in adulthood. Finally, neither post-weanling nor peri-pubertal dcc-heterozygous mice demonstrate the blunted amphetamine-induced locomotor response observed in adulthood. Thus, these findings show that the 'protective' dcc phenotype has a post-pubertal emergence and indicate that DCC may play a role in the normal maturation of the mesocorticolimbic DA system.
Collapse
Affiliation(s)
- Alanna Grant
- Departments of Psychiatry and Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|
45
|
Loss of dendrite stabilization by the Abl-related gene (Arg) kinase regulates behavioral flexibility and sensitivity to cocaine. Proc Natl Acad Sci U S A 2009; 106:16859-64. [PMID: 19805386 DOI: 10.1073/pnas.0902286106] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Adolescence is characterized by increased vulnerability to developing neuropsychiatric disorders and involves a period of prefrontal cortical dendritic refinement and synaptic pruning that culminates in cytoskeletal stabilization in adulthood. The Abl-related gene (Arg) acts through p190RhoGAP to inhibit the RhoA GTPase and stabilize cortical dendritic arbors beginning in adolescence. Cortical axons, dendrites, and synapses develop normally in Arg-deficient (arg(-/-)) mice, but adult dendrites destabilize and regress; thus, arg(-/-) mice present a model of adolescent-onset dendritic simplification. We show that arg(-/-) mice are impaired in a reversal task and that deficits are grossly exacerbated by low-dose cocaine administration. Although ventral prefrontal dopamine D2 receptor levels predict "perseverative" error counts in wild-type mice, no such relationship is found in arg(-/-) mice. Moreover, arg(-/-) mice are insensitive to the disruptive effects of the D2/D3 antagonist haloperidol in reversal but show normal sensitivity to its locomotor-depressant actions. Arg deficiency and orbitofrontal cortical Arg inhibition via STI-571 infusion also enhance the psychomotor stimulant actions of cocaine. These findings provide evidence that stabilization of dendritic structure beginning in adolescence is critical for the development of adaptive and flexible behavior after cocaine exposure.
Collapse
|
46
|
Boyce PJ, Finlay JM. Extracellular dopamine and norepinephrine in the developing rat prefrontal cortex: transient effects of early partial loss of dopamine. Brain Res Bull 2009; 79:104-110. [PMID: 19320060 DOI: 10.1016/j.brainresbull.2009.01.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Early developmental abnormalities affecting mesocortical dopamine (DA) neurons may result in later functional deficits that play a role in the emergence of psychiatric illness in adolescence/early adulthood. Little is known about the functional maturation of these neurons under either normal or abnormal conditions. In the present study, 6-hydroxydopamine was infused into the rat medial prefrontal cortex (mPFC) on postnatal day (PN) 12-14. On PN30-35, 45-50, and 60-65, mPFC extracellular DA and norepinephrine (NE) concentrations were monitored in intact and lesioned rats using in vivo microdialysis. Extracellular DA and NE concentrations in the intact mPFC remain fairly stable across development; one exception being a trend for acute tailshock-evoked DA concentrations to increase as a function of age. Lesioned rats sustained a persistent (approximately 50%) decrease in mPFC tissue DA concentrations. Tailshock-evoked increases in mPFC extracellular DA were attenuated in lesioned rats tested on PN30-35, but not PN45-50 or 60-65. Basal and evoked extracellular NE was unaffected in lesioned rats tested at any age, despite a persistent (approximately 25%) decrease in tissue NE content. Horizontal locomotor activity was also assessed in the present study. Results of previous studies suggest this behavior is modulated by mesoprefrontal DA neurons. Although not significant, acute tailshock- and acute amphetamine-evoked horizontal locomotor activity tended to be attenuated in lesioned rats tested on PN30-35 and augmented in lesioned rats tested on PN60-65. The present data suggest that early partial loss of mesoprefrontal DA nerve terminals, resulting in a persistent decrease in tissue DA concentrations, is unlikely to result in persistent alterations in local DA release.
Collapse
Affiliation(s)
- Patricia J Boyce
- Department of Psychology, Western Washington University, Bellingham, WA 98225-9089, United States
| | | |
Collapse
|
47
|
Berman SM, Kuczenski R, McCracken JT, London ED. Potential adverse effects of amphetamine treatment on brain and behavior: a review. Mol Psychiatry 2009; 14:123-42. [PMID: 18698321 PMCID: PMC2670101 DOI: 10.1038/mp.2008.90] [Citation(s) in RCA: 165] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Revised: 07/03/2008] [Accepted: 07/18/2008] [Indexed: 01/09/2023]
Abstract
Amphetamine stimulants have been used medically since early in the twentieth century, but they have a high abuse potential and can be neurotoxic. Although they have long been used effectively to treat attention deficit hyperactivity disorder (ADHD) in children and adolescents, amphetamines are now being prescribed increasingly as maintenance therapy for ADHD and narcolepsy in adults, considerably extending the period of potential exposure. Effects of prolonged stimulant treatment have not been fully explored, and understanding such effects is a research priority. Because the pharmacokinetics of amphetamines differ between children and adults, reevaluation of the potential for adverse effects of chronic treatment of adults is essential. Despite information on the effects of stimulants in laboratory animals, profound species differences in susceptibility to stimulant-induced neurotoxicity underscore the need for systematic studies of prolonged human exposure. Early amphetamine treatment has been linked to slowing in height and weight growth in some children. Because the number of prescriptions for amphetamines has increased several fold over the past decade, an amphetamine-containing formulation is the most commonly prescribed stimulant in North America, and it is noteworthy that amphetamines are also the most abused prescription medications. Although early treatment does not increase risk for substance abuse, few studies have tracked the compliance and usage profiles of individuals who began amphetamine treatment as adults. Overall, there is concern about risk for slowed growth in young patients who are dosed continuously, and for substance abuse in patients first medicated in late adolescence or adulthood. Although most adult patients also use amphetamines effectively and safely, occasional case reports indicate that prescription use can produce marked psychological adverse events, including stimulant-induced psychosis. Assessments of central toxicity and adverse psychological effects during late adulthood and senescence of adults who receive prolonged courses of amphetamine treatment are warranted. Finally, identification of the biological factors that confer risk and those that offer protection is also needed to better specify the parameters of safe, long-term, therapeutic administration of amphetamines to adults.
Collapse
Affiliation(s)
- S M Berman
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90024-1759, USA
| | | | | | | |
Collapse
|
48
|
Harrison LM, Lahoste GJ, Ruskin DN. Ontogeny and dopaminergic regulation in brain of Ras homolog enriched in striatum (Rhes). Brain Res 2008; 1245:16-25. [PMID: 18929545 PMCID: PMC2615551 DOI: 10.1016/j.brainres.2008.09.066] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Revised: 09/12/2008] [Accepted: 09/23/2008] [Indexed: 11/20/2022]
Abstract
Rhes is one of several signaling molecules preferentially expressed in the striatum. This GTP-binding protein affects dopamine-mediated signaling and behavior. Denervating the striatum of its dopaminergic inputs in adulthood reduces rhes mRNA expression. Here we show that dopamine depletion in adult rats by 6-hydroxydopamine caused a significant decrease in striatal Rhes protein levels as measured by Western blotting. The role of dopamine input on rhes mRNA induction during ontogeny was also examined. Rhes mRNA was measured on postnatal days 4, 6, 8, 10, 15, and 24 with in situ hybridization to determine its normal ontogeny. Signal in striatum was detectable, but very low, on postnatal day 4 and increased gradually to peak levels at days 15 and 24. Outside of the striatum, rhes mRNA was expressed at high levels in hippocampus and cerebellum during the postnatal period. Hippocampal signal was initially highest in CA3 and dentate gyrus, but shifted to higher expression in CA1 by the late postnatal period. Several other nuclei showed low levels of rhes mRNA during ontogeny. Depletion of dopamine by 6-hydroxydopamine injection on postnatal day 4 did not affect the ontogenetic development of rhes mRNA, such that expression did not differ statistically in lesioned versus vehicle-treated animals tested in adulthood. These findings suggest that although dopamine input is not necessary for the ontogenetic development of rhes mRNA expression, changes in both rhes mRNA and Rhes protein are integral components of the response of the adult striatum to dopamine depletion.
Collapse
Affiliation(s)
- Laura M Harrison
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, New Orleans, LA 70112, USA.
| | | | | |
Collapse
|
49
|
Paz RD, Tardito S, Atzori M, Tseng KY. Glutamatergic dysfunction in schizophrenia: from basic neuroscience to clinical psychopharmacology. Eur Neuropsychopharmacol 2008; 18:773-86. [PMID: 18650071 PMCID: PMC2831778 DOI: 10.1016/j.euroneuro.2008.06.005] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Revised: 05/27/2008] [Accepted: 06/17/2008] [Indexed: 01/02/2023]
Abstract
The underlying cellular mechanisms leading to frontal cortical hypofunction (i.e., hypofrontality) in schizophrenia remain unclear. Both hypoactive and hyperreactive prefrontal cortical (PFC) states have been reported in schizophrenia patients. Recent proton magnetic resonance spectroscopy studies revealed that antipsychotic-naïve patients with first psychotic episode exhibit a hyperactive PFC. Conversely, PFC activity seems to be diminished in patients chronically exposed to conventional antipsychotic treatments, an effect that could reflect the therapeutic action as well as some of the impairing side effects induced by long-term blockade of dopamine transmission. In this review, we will provide an evolving picture of the pathophysiology of schizophrenia moving from dopamine to a more glutamatergic-centered hypothesis. We will discuss how alternative antipsychotic strategies may emerge by using drugs that reduce excessive glutamatergic response without altering the balance of synaptic and extrasynaptic normal glutamatergic neurotransmission. Preclinical studies indicate that acamprosate, a FDA approved drug for relapse prevention in detoxified alcoholic patients, reduces the glutamatergic hyperactivity triggered by ethanol withdrawal without depressing normal glutamatergic transmission. Whether this effect is mediated by a direct modulation of NMDA receptors or by antagonism of metabotropic glutamate receptor remains to be determined. We hypothesize that drugs with similar pharmacological actions to acamprosate may provide a better and safer approach to reverse psychotic symptoms and cognitive deficits without altering the balance of excitation and inhibition of the corticolimbic dopamine-PFC system. It is predicted that schizophrenia patients treated with acamprosate-like compounds will not exhibit progressive cortical atrophy associated with the anti-dopaminergic effect of classical antipsychotic exposure.
Collapse
Affiliation(s)
- Rodrigo D. Paz
- Departamento de Psiquiatría y Neurociencias, Universidad Diego Portales, Santiago, Chile
- Instituto Psiquiátrico José Horwitz Barak, Santiago, Chile
| | - Sonia Tardito
- Instituto Psiquiátrico José Horwitz Barak, Santiago, Chile
| | - Marco Atzori
- University of Texas at Dallas, School for Behavioral and Brain Sciences, Richardson, Texas, USA
| | - Kuei Y. Tseng
- Department of Cellular & Molecular Pharmacology, RFUMS/The Chicago Medical School, North Chicago, Illinois, USA
| |
Collapse
|
50
|
Faria RR, Lima Rueda AV, Sayuri C, Soares SL, Malta MB, Carrara-Nascimento PF, da Silva Alves A, Marcourakis T, Yonamine M, Scavone C, Giorgetti Britto LR, Camarini R. Environmental modulation of ethanol-induced locomotor activity: Correlation with neuronal activity in distinct brain regions of adolescent and adult Swiss mice. Brain Res 2008; 1239:127-40. [PMID: 18789904 DOI: 10.1016/j.brainres.2008.08.056] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2008] [Revised: 08/18/2008] [Accepted: 08/20/2008] [Indexed: 10/21/2022]
Abstract
Drug abuse is a concerning health problem in adults and has been recognized as a major problem in adolescents. Induction of immediate-early genes (IEG), such as c-Fos or Egr-1, is used to identify brain areas that become activated in response to various stimuli, including addictive drugs. It is known that the environment can alter the response to drugs of abuse. Accordingly, environmental cues may trigger drug-seeking behavior when the drug is repeatedly administered in a given environment. The goal of this study was first to examine for age differences in context-dependent sensitization and then evaluate IEG expression in different brain regions. For this, groups of mice received i.p. ethanol (2.0 g/kg) or saline in the test apparatus, while other groups received the solutions in the home cage, for 15 days. One week after this treatment phase, mice were challenged with ethanol injection. Acutely, ethanol increased both locomotor activity and IEG expression in different brain regions, indistinctly, in adolescent and adult mice. However, adults exhibited a typical context-dependent behavioral sensitization following repeated ethanol treatment, while adolescent mice presented gradually smaller locomotion across treatment, when ethanol was administered in a paired regimen with environment. Conversely, ethanol-treated adolescents expressed context-independent behavioral sensitization. Overall, repeated ethanol administration desensitized IEG expression in both adolescent and adult mice, but this effect was greatest in the nucleus accumbens and prefrontal cortex of adolescents treated in the context-dependent paradigm. These results suggest developmental differences in the sensitivity to the conditioned and unconditioned locomotor effects of ethanol.
Collapse
Affiliation(s)
- Rulian Ricardo Faria
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, 05508-900, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|