1
|
Topchiy I, Mohbat J, Folorunso OO, Wang ZZ, Lazcano-Etchebarne C, Engin E. GABA system as the cause and effect in early development. Neurosci Biobehav Rev 2024; 161:105651. [PMID: 38579901 PMCID: PMC11081854 DOI: 10.1016/j.neubiorev.2024.105651] [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: 01/03/2024] [Revised: 03/05/2024] [Accepted: 04/01/2024] [Indexed: 04/07/2024]
Abstract
GABA is the primary inhibitory neurotransmitter in the adult brain and through its actions on GABAARs, it protects against excitotoxicity and seizure activity, ensures temporal fidelity of neurotransmission, and regulates concerted rhythmic activity of neuronal populations. In the developing brain, the development of GABAergic neurons precedes that of glutamatergic neurons and the GABA system serves as a guide and framework for the development of other brain systems. Despite this early start, the maturation of the GABA system also continues well into the early postnatal period. In this review, we organize evidence around two scenarios based on the essential and protracted nature of GABA system development: 1) disruptions in the development of the GABA system can lead to large scale disruptions in other developmental processes (i.e., GABA as the cause), 2) protracted maturation of this system makes it vulnerable to the effects of developmental insults (i.e., GABA as the effect). While ample evidence supports the importance of GABA/GABAAR system in both scenarios, large gaps in existing knowledge prevent strong mechanistic conclusions.
Collapse
Affiliation(s)
- Irina Topchiy
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA
| | - Julie Mohbat
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA; School of Life Sciences, Ecole Polytechnique Federale de Lausanne, Lausanne CH-1015, Switzerland
| | - Oluwarotimi O Folorunso
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA
| | - Ziyi Zephyr Wang
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA
| | | | - Elif Engin
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
2
|
McCarthy DM, Spencer TJ, Bhide PG. Preclinical Models of Attention Deficit Hyperactivity Disorder: Neurobiology, Drug Discovery, and Beyond. J Atten Disord 2024; 28:880-894. [PMID: 38084074 DOI: 10.1177/10870547231215286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
OBJECTIVE We offer an overview of ADHD research using mouse models of nicotine exposure. METHOD Nicotine exposure of C57BL/6 or Swiss Webster mice occurred during prenatal period only or during the prenatal and the pre-weaning periods. Behavioral, neuroanatomical and neurotransmitter assays were used to investigate neurobiological mechanisms of ADHD and discover candidate ADHD medications. RESULTS Our studies show that norbinaltorphimine, a selective kappa opioid receptor antagonist is a candidate novel non-stimulant ADHD treatment and that a combination of methylphenidate and naltrexone has abuse deterrent potential with therapeutic benefits for ADHD. Other studies showed transgenerational transmission of ADHD-associated behavioral traits and demonstrated that interactions between untreated ADHD and repeated mild traumatic brain injury produced behavioral traits not associated with either condition alone. CONCLUSION Preclinical models contribute to novel insights into ADHD neurobiology and are valuable tools for drug discovery and translation to benefit humans with ADHD.
Collapse
Affiliation(s)
| | - Thomas J Spencer
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Pradeep G Bhide
- Florida State University College of Medicine, Tallahassee, FL, USA
| |
Collapse
|
3
|
Rudibaugh TT, Stuppy SR, Keung AJ. Reactive Oxygen Species Mediate Transcriptional Responses to Dopamine and Cocaine in Human Cerebral Organoids. Int J Mol Sci 2023; 24:16474. [PMID: 38003664 PMCID: PMC10671319 DOI: 10.3390/ijms242216474] [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: 10/24/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Dopamine signaling in the adult ventral forebrain regulates behavior, stress response, and memory formation and in neurodevelopment regulates neural differentiation and cell migration. Excessive dopamine levels, including those due to cocaine use in utero and in adults, could lead to long-term adverse consequences. The mechanisms underlying both homeostatic and pathological changes remain unclear, in part due to the diverse cellular responses elicited by dopamine and the reliance on animal models that exhibit species-specific differences in dopamine signaling. In this study, we use the human-derived ventral forebrain organoid model of Xiang-Tanaka and characterize their response to cocaine or dopamine. We explore dosing regimens of dopamine or cocaine to simulate acute or chronic exposure. We then use calcium imaging, cAMP imaging, and bulk RNA-sequencing to measure responses to cocaine or dopamine exposure. We observe an upregulation of inflammatory pathways in addition to indicators of oxidative stress following exposure. Using inhibitors of reactive oxygen species (ROS), we then show ROS to be necessary for multiple transcriptional responses of cocaine exposure. These results highlight novel response pathways and validate the potential of cerebral organoids as in vitro human models for studying complex biological processes in the brain.
Collapse
Affiliation(s)
| | | | - Albert J. Keung
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27606, USA; (T.T.R.); (S.R.S.)
| |
Collapse
|
4
|
Rudibaugh TT, Keung AJ. Reactive Oxygen Species Mediate Transcriptional Responses to Dopamine and Cocaine in Human Cerebral Organoids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.13.544782. [PMID: 37398046 PMCID: PMC10312668 DOI: 10.1101/2023.06.13.544782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Dopamine signaling in the adult ventral forebrain regulates behavior, stress response, and memory formation and in neurodevelopment regulates neural differentiation and cell migration. Excessive dopamine levels including due to cocaine use both in utero and in adults could lead to long-term adverse consequences. The mechanisms underlying both homeostatic and pathological changes remain unclear, partly due to the diverse cellular responses elicited by dopamine and the reliance on animal models that exhibit species-specific differences in dopamine signaling. To address these limitations, 3-D cerebral organoids have emerged as human-derived models, recapitulating salient features of human cell signaling and neurodevelopment. Organoids have demonstrated responsiveness to external stimuli, including substances of abuse, making them valuable investigative models. In this study we utilize the Xiang-Tanaka ventral forebrain organoid model and characterize their response to acute and chronic dopamine or cocaine exposure. The findings revealed a robust immune response, novel response pathways, and a potential critical role for reactive oxygen species (ROS) in the developing ventral forebrain. These results highlight the potential of cerebral organoids as in vitro human models for studying complex biological processes in the brain.
Collapse
Affiliation(s)
- Thomas T. Rudibaugh
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27606
| | - Albert J. Keung
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27606
| |
Collapse
|
5
|
Maternal Hyperhomocysteinemia Disturbs the Mechanisms of Embryonic Brain Development and Its Maturation in Early Postnatal Ontogenesis. Cells 2023; 12:cells12010189. [PMID: 36611982 PMCID: PMC9818313 DOI: 10.3390/cells12010189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/05/2023] Open
Abstract
Maternal hyperhomocysteinemia causes the disruption of placental blood flow and can lead to serious disturbances in the formation of the offspring's brain. In the present study, the effects of prenatal hyperhomocysteinemia (PHHC) on the neuronal migration, neural tissue maturation, and the expression of signaling molecules in the rat fetal brain were described. Maternal hyperhomocysteinemia was induced in female rats by per os administration of 0.15% aqueous methionine solution in the period of days 4-21 of pregnancy. Behavioral tests revealed a delay in PHHC male pups maturing. Ultrastructure of both cortical and hippocampus tissue demonstrated the features of the developmental delay. PHHC was shown to disturb both generation and radial migration of neuroblasts into the cortical plate. Elevated Bdnf expression, together with changes in proBDNF/mBDNF balance, might affect neuronal cell viability, positioning, and maturation in PHHC pups. Reduced Kdr gene expression and the content of SEMA3E might lead to impaired brain development. In the brain tissue of E20 PHHC fetuses, the content of the procaspase-8 was decreased, and the activity level of the caspase-3 was increased; this may indicate the development of apoptosis. PHHC disturbs the mechanisms of early brain development leading to a delay in brain tissue maturation and formation of the motor reaction of pups.
Collapse
|
6
|
McCarthy DM, Zhang L, Wilkes BJ, Vaillancourt DE, Biederman J, Bhide PG. Nicotine and the developing brain: Insights from preclinical models. Pharmacol Biochem Behav 2022; 214:173355. [PMID: 35176350 PMCID: PMC9063417 DOI: 10.1016/j.pbb.2022.173355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 11/26/2022]
Abstract
Use of tobacco products during pregnancy is associated with increased risk for neurodevelopmental disorders in the offspring. Preclinical models of developmental nicotine exposure have offered valuable insights into the neurobiology of nicotine's effects on the developing brain and demonstrated lasting effects of developmental nicotine exposure on brain structure, neurotransmitter signaling and behavior. These models have facilitated discovery of novel compounds as candidate treatments for attention deficit hyperactivity disorder, a neurodevelopmental disorder associated with prenatal nicotine exposure. Using these models the significance of heritability of behavioral phenotypes from the nicotine-exposed pregnant female or adult male to multiple generations of descendants has been demonstrated. Finally, research using the preclinical models has demonstrated synergistic interactions between developmental nicotine exposure and repetitive mild traumatic brain injury that contribute to "worse" outcomes from the injury in individuals with attention deficit hyperactivity disorder associated with developmental nicotine exposure.
Collapse
Affiliation(s)
- Deirdre M McCarthy
- Biomedical Sciences, Florida State University, College of Medicine, Tallahassee, FL 32306, United States of America
| | - Lin Zhang
- Biomedical Sciences, Florida State University, College of Medicine, Tallahassee, FL 32306, United States of America
| | - Bradley J Wilkes
- Laboratory for Rehabilitation Neuroscience, Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL 32611, United States of America
| | - David E Vaillancourt
- Laboratory for Rehabilitation Neuroscience, Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL 32611, United States of America
| | - Joseph Biederman
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States of America
| | - Pradeep G Bhide
- Biomedical Sciences, Florida State University, College of Medicine, Tallahassee, FL 32306, United States of America.
| |
Collapse
|
7
|
Qiu X, Chen H, Feng D, Dong W. [G-protein coupled receptor Smo positively regulates proliferation and migration of adult neural stem cells in vitro]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:1588-1592. [PMID: 34755677 DOI: 10.12122/j.issn.1673-4254.2021.10.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the role of G-protein coupled receptor Smoothened (Smo) in regulating proliferation and migration of adult neural stem cells (ANSCs) and explore the underlying mechanism. METHODS Cultured ANSCs were treated with purmorphamine (PM, an agonist of Smo) or cyclopamine (CPM, an inhibitor of Smo), and the changes in cell proliferation migration abilities were assessed using cell counting kit-8 (CCK8) assay and wound healing assay, respectively. The mRNA expressions of membrane receptor Patched 1 (Ptch1), Smo, glioma-associated oncogene homolog 1 (Gli1), axon guidance cue slit1 (Slit1) and brain-derived neurotrophic factor (BDNF) in the treated cells were detected using real-time quantitative PCR (RT-PCR). RESULTS PM significantly promoted the proliferation (P < 0.01) and migration of ANSCs (P < 0.01), and up-regulated the mRNA expressions of Ptch1, Smo, Gli1, Slit1 and BDNF. Treatment with CPM significantly inhibited the proliferation and migration of ANSCs. CONCLUSION Modulating Smo activity can positively regulate the proliferation and migration of ANSCs possibly by regulating the expressions of BDNF and Slit1.
Collapse
Affiliation(s)
- X Qiu
- Experiment Teaching and Administration Center, Southern Medical University, Guangzhou 510515, China
| | - H Chen
- Department of Neurosurgery, Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - D Feng
- Institute of Oncology, Southern Medical University, Guangzhou 510515, China
| | - W Dong
- Experiment Teaching and Administration Center, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
8
|
Martin MM, McCarthy DM, Schatschneider C, Trupiano MX, Jones SK, Kalluri A, Bhide PG. Effects of Developmental Nicotine Exposure on Frontal Cortical GABA-to-Non-GABA Neuron Ratio and Novelty-Seeking Behavior. Cereb Cortex 2021; 30:1830-1842. [PMID: 31599922 DOI: 10.1093/cercor/bhz207] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Cigarette smoking during pregnancy is a major public health concern, resulting in detrimental health effects in the mother and her offspring. The adverse behavioral consequences for children include increased risk for attention deficit hyperactivity disorder, working memory deficits, epilepsy, novelty-seeking, and risk-taking behaviors. Some of these behavioral conditions are consistent with an imbalance in frontal cortical excitatory (glutamate) and inhibitory (GABA) neurotransmitter signaling. We used a GAD67-GFP knock-in mouse model to examine if developmental nicotine exposure alters frontal cortical GABA neuron numbers, GABA-to-non-GABA neuron ratio and behavioral phenotypes. Female mice were exposed to nicotine (100 or 200 μg/mL) in drinking water beginning 3 weeks prior to breeding and until 3 weeks postpartum. Male and female offspring were examined beginning at 60 days of age. The nicotine exposure produced dose-dependent decreases in GABA-to-non-GABA neuron ratios in the prefrontal and medial prefrontal cortices without perturbing the intrinsic differences in cortical thickness and laminar distribution of GABA or non-GABA neurons between these regions. A significant increase in exploratory behavior and a shift toward "approach" in the approach-avoidance paradigm were also observed. Thus, developmental nicotine exposure shifts the cortical excitation-inhibition balance toward excitation and produces behavioral changes consistent with novelty-seeking behavior.
Collapse
Affiliation(s)
- Melissa M Martin
- Center for Brain Repair, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306-4300, USA
| | - Deirdre M McCarthy
- Center for Brain Repair, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306-4300, USA
| | - Chris Schatschneider
- Department of Psychology, Florida State University, Tallahassee, FL 32306-4300, USA
| | - Mia X Trupiano
- Center for Brain Repair, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306-4300, USA
| | - Sara K Jones
- Center for Brain Repair, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306-4300, USA
| | - Aishani Kalluri
- Center for Brain Repair, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306-4300, USA
| | - Pradeep G Bhide
- Center for Brain Repair, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306-4300, USA
| |
Collapse
|
9
|
A Progressive Loss of phosphoSer138-Profilin Aligns with Symptomatic Course in the R6/2 Mouse Model of Huntington's Disease: Possible Sex-Dependent Signaling. Cell Mol Neurobiol 2020; 42:871-888. [PMID: 33108594 PMCID: PMC8891113 DOI: 10.1007/s10571-020-00984-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 10/16/2020] [Indexed: 12/20/2022]
Abstract
The R6/2 transgenic mouse model of Huntington’s disease (HD) carries several copies of exon1 of the huntingtin gene that contains a highly pathogenic 120 CAG-repeat expansion. We used kinome analysis to screen for kinase activity patterns in neural tissues from wildtype (WT) and R6/2 mice at a pre-symptomatic (e.g., embryonic) and symptomatic (e.g., between 3 and 10 weeks postnatal) time points. We identified changes in several signaling cascades, for example, the Akt/FoxO3/CDK2, mTOR/ULK1, and RAF/MEK/CREB pathways. We also identified the Rho-Rac GTPase cascade that contributes to cytoskeleton organization through modulation of the actin-binding proteins, cofilin and profilin. Immunoblotting revealed higher levels of phosphoSer138-profilin in embryonic R6/2 mouse samples (cf. WT mice) that diminish progressively and significantly over the postnatal, symptomatic course of the disease. We detected sex- and genotype-dependent patterns in the phosphorylation of actin-regulators such a ROCK2, PAK, LIMK1, cofilin, and SSH1L, yet none of these aligned consistently with the changing levels of phosphoSer138-profilin. This could be reflecting an imbalance in the sequential influences these regulators are known to exert on actin signaling. The translational potential of these observations was inferred from preliminary observations of changes in LIMK-cofilin signaling and loss of neurite integrity in neural stem cells derived from an HD patient (versus a healthy control). Our observations suggest that a pre-symptomatic, neurodevelopmental onset of change in the phosphorylation of Ser138-profilin, potentially downstream of distinct signaling changes in male and female mice, could be contributing to cytoskeletal phenotypes in the R6/2 mouse model of HD pathology.
Collapse
|
10
|
Rivera P, Aranda J, Alén F, Vargas A, Serrano A, Pavón FJ, Orio L, Rubio L, Moratalla R, de Fonseca FR, Suárez J. Sex-specific behavioral and neurogenic responses to cocaine in mice lacking and blocking dopamine D1 or dopamine D2 receptors. J Comp Neurol 2020; 529:1724-1742. [PMID: 33047300 DOI: 10.1002/cne.25052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 07/21/2020] [Accepted: 10/03/2020] [Indexed: 12/22/2022]
Abstract
Adult neurogenesis in rodents is modulated by dopaminergic signaling and inhibited by cocaine. However, the sex-specific role of dopamine D1 and D2 receptors (D1R, D2R) in the deleterious effect of cocaine on adult neurogenesis has not been described yet. Here, we explored sex differences in (a) cell proliferation (5'-bromo-2'-deoxyuridine [BrdU]), (b) neural precursor (nestin), (c) neuronal phenotype (BrdU/β3-tubulin), and (d) neuronal maturity (NeuN) in the subventricular zone (SVZ) of the lateral ventricles and striatum of mice with genetic deletion (D1-/- , D2-/- ) or pharmacological blockage (SCH23390: 0.1 mg/kg/day/5 days; Raclopride: 0.3 mg/kg/day/5 days) of D1R and D2R, and treated (10 mg/kg/day/5 days) and then challenged (5 mg/kg, 48 hr later) with cocaine. Results indicated that hyperactivity responses to cocaine were absent in D1-/- mice and reduced in SCH23390-treated mice. Activity responses to cocaine were reduced in D2-/- males, but absent in D2-/- females and increased in Raclopride-treated females. D1R deletion blocked the deleterious effect of cocaine on SVZ cell proliferation in males. Cocaine-exposed D1-/- males also had reduced neuronal phenotype of SVZ newborn cells and increased striatal neuronal maturity. D2-/- mice had lower proliferative and neural precursor responses. Cocaine in D2-/- females or coadministered with Raclopride in wild-type females improved SVZ cell proliferation, an effect that positively correlated with plasma brain-derived neurotrophic factor (BDNF) concentrations. In conclusion, the sex-specific D1R and D2R signaling on SVZ cell proliferation, neural progenitor and neuronal maturity is differentially perturbed by cocaine, and BDNF may be required to link D2R to neuroplasticity in cocaine addiction in females.
Collapse
Affiliation(s)
- Patricia Rivera
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, Universidad de Málaga, Málaga, Spain
| | - Jesús Aranda
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, Universidad de Málaga, Málaga, Spain
| | - Francisco Alén
- Departamento de Psicobiología, Universidad Complutense de Madrid, Madrid, Spain
| | - Antonio Vargas
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, Universidad de Málaga, Málaga, Spain
| | - Antonia Serrano
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, Universidad de Málaga, Málaga, Spain
| | - Francisco Javier Pavón
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, Universidad de Málaga, Málaga, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain; and UGC Corazón, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain
| | - Laura Orio
- Departamento de Psicobiología, Universidad Complutense de Madrid, Madrid, Spain
| | - Leticia Rubio
- Departamento de Anatomía Humana y Medicina Legal, Universidad de Málaga, Málaga, Spain
| | - Rosario Moratalla
- Instituto Cajal de Madrid, Consejo Superior de Investigaciones Científica, Madrid, Spain
| | - Fernando Rodríguez de Fonseca
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, Universidad de Málaga, Málaga, Spain
| | - Juan Suárez
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, Universidad de Málaga, Málaga, Spain
| |
Collapse
|
11
|
Pasquet N, Douceau S, Naveau M, Lesept F, Louessard M, Lebouvier L, Hommet Y, Vivien D, Bardou I. Tissue-Type Plasminogen Activator Controlled Corticogenesis Through a Mechanism Dependent of NMDA Receptors Expressed on Radial Glial Cells. Cereb Cortex 2020; 29:2482-2498. [PMID: 29878094 DOI: 10.1093/cercor/bhy119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Indexed: 01/24/2023] Open
Abstract
Modifications of neuronal migration during development, including processes that control cortical lamination are associated with functional deficits at adult stage. Here, we report for the first time that the lack of the serine protease tissue-type Plasminogen Activator (tPA), previously characterized as a neuromodulator and a gliotransmitter, leads to an altered cortical lamination in adult. This results in a neuronal migration defect of tPA deficient neurons which are stopped in the intermediate zone at E16. This phenotype is rescued by re-expressing a wild-type tPA in cortical neurons at E14 but not by a tPA that cannot interact with NMDAR. We thus hypothetized that the tPA produced by cortical neuronal progenitors can control their own radial migration through a mechanism dependent of NMDAR expressed at the surface of radial glial cells (RGC). Accordingly, conditional deletion of tPA in neuronal progenitors at E14 or overexpression of a dominant-negative NMDAR that cannot bind tPA in RGC also delayed neuronal migration. Moreover, the lack of tPA lead to an impaired maturation and orientation of RGC. These data provide the first demonstration that the neuronal serine protease tPA is an actor of a proper corticogenesis by its ability to control NMDAR signaling in RGC.
Collapse
Affiliation(s)
- Nolwenn Pasquet
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Sara Douceau
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Mickael Naveau
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Flavie Lesept
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Morgane Louessard
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Laurent Lebouvier
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Yannick Hommet
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Denis Vivien
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France.,CHU Caen, Clinical Research Department, Caen University Hospital, Caen, France
| | - Isabelle Bardou
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| |
Collapse
|
12
|
Hwang HM, Ku RY, Hashimoto-Torii K. Prenatal Environment That Affects Neuronal Migration. Front Cell Dev Biol 2019; 7:138. [PMID: 31380373 PMCID: PMC6652208 DOI: 10.3389/fcell.2019.00138] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/08/2019] [Indexed: 11/22/2022] Open
Abstract
Migration of neurons starts in the prenatal period and continues into infancy. This developmental process is crucial for forming a proper neuronal network, and the disturbance of this process results in dysfunction of the brain such as epilepsy. Prenatal exposure to environmental stress, including alcohol, drugs, and inflammation, disrupts neuronal migration and causes neuronal migration disorders (NMDs). In this review, we summarize recent findings on this topic and specifically focusing on two different modes of migration, radial, and tangential migration during cortical development. The shared mechanisms underlying the NMDs are discussed by comparing the molecular changes in impaired neuronal migration under exposure to different types of prenatal environmental stress.
Collapse
Affiliation(s)
- Hye M Hwang
- Center for Neuroscience Research, Children's National Medical Center, The Children's Research Institute, Washington, DC, United States.,The Institute for Biomedical Sciences, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Ray Y Ku
- Center for Neuroscience Research, Children's National Medical Center, The Children's Research Institute, Washington, DC, United States
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, Children's National Medical Center, The Children's Research Institute, Washington, DC, United States.,Departments of Pediatrics, and Pharmacology & Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| |
Collapse
|
13
|
Yaw AM, Prosser RA, Jones PC, Garcia BJ, Jacobson DA, Glass JD. Epigenetic effects of paternal cocaine on reward stimulus behavior and accumbens gene expression in mice. Behav Brain Res 2019; 367:68-81. [PMID: 30910707 DOI: 10.1016/j.bbr.2019.02.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/08/2019] [Accepted: 02/25/2019] [Indexed: 12/23/2022]
Abstract
Paternal cocaine use causes phenotypic alterations in offspring behavior and associated neural processing. In rodents, changes in first generation (F1) offspring include drug reward behavior, circadian timing, and anxiety responses. This study, utilizing a murine (C57BL/6J) oral cocaine model, examines the effects of paternal cocaine exposure on fundamental characteristics of offspring reward responses, including: 1) the extent of cocaine-induced effects after different durations of sire drug withdrawal; 2) sex- and drug-dependent differences in F1 reward preference; 3) effects on second generation (F2) cocaine preference; and 4) corresponding changes in reward area (nucleus accumbens) mRNA expression. We demonstrate that paternal cocaine intake over a single ˜40-day spermatogenic cycle significantly decreased cocaine (but not ethanol or sucrose) preference in a sex-specific manner in F1 mice from sires mated 24 h after drug withdrawal. However, F1 offspring of sires bred 4 months after withdrawal did not exhibit altered cocaine preference. Altered cocaine preference also was not observed in F2's. RNASeq analyses of F1 accumbens tissue revealed changes in gene expression in male offspring of cocaine-exposed sires, including many genes not previously linked to cocaine addiction. Enrichment analyses highlight genes linked to CNS development, synaptic signaling, extracellular matrix, and immune function. Expression correlation analyses identified a novel target, Fam19a4, that may negatively regulate many genes in the accumbens, including genes already identified in addiction. Collectively, these results reveal that paternal cocaine effects in F1 offspring may involve temporally limited epigenetic germline effects and identify new genetic targets for addiction research.
Collapse
Affiliation(s)
- Alexandra M Yaw
- School of Biomedical Sciences, Kent State Univ., Kent, OH, 44242, United States
| | - Rebecca A Prosser
- Dept. of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, 37996, United States; NeuroNET Research Center, University of Tennessee, Knoxville, TN, 37996, United States
| | - Piet C Jones
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37830, United States; Bredesen Center for Interdisciplinary Research, University of Tennessee, Knoxville, TN, 37996, United States
| | - Benjamin J Garcia
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37830, United States
| | - Daniel A Jacobson
- NeuroNET Research Center, University of Tennessee, Knoxville, TN, 37996, United States; Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37830, United States; Bredesen Center for Interdisciplinary Research, University of Tennessee, Knoxville, TN, 37996, United States; Department of Psychology, University of Tennessee, Knoxville, TN, 37996, United States
| | - J David Glass
- School of Biomedical Sciences, Kent State Univ., Kent, OH, 44242, United States.
| |
Collapse
|
14
|
Maternal methamphetamine exposure causes cognitive impairment and alteration of neurodevelopment-related genes in adult offspring mice. Neuropharmacology 2018; 140:25-34. [PMID: 30048643 DOI: 10.1016/j.neuropharm.2018.07.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/05/2018] [Accepted: 07/22/2018] [Indexed: 12/26/2022]
Abstract
Prenatal drug exposure altered cognitive function in individuals, and may also impact their offspring's susceptibility to cognitive impairment. The high incidence of methamphetamine (METH) abuse among adolescents and women of childbearing age elevates the importance to determine the influence of maternal METH exposure on cognitive functions in the descendants. We hypothesized that maternal METH exposure affects cognitive behavior in offspring mice by disrupting gene expression associated with neural development. Here, female C57BL/6 mice were exposed to intermittent escalating doses of METH or saline from adolescence to adulthood, and then continued through pregnancy. Interestingly, male but not female offspring exhibited impaired short-term recognition memory and long-term spatial memory retention in novel object recognition and Morris water maze test respectively. Additionally, maternal METH exposure altered neurodevelopmental genes in both male and female offspring, and 12 differentially expressed genes between male and female were observed in the HPC and NAc regions. These differentially expressed genes are involved in neurogenesis, axon guidance, neuron migration and synapse of neural development circuits. Our observations suggest that maternal METH exposure induced differential expression patterns of neurodevelopment-related genes in the HPC and NAc of male and female mice, which may underlie the different cognitive behavior phenotypes in both genders.
Collapse
|
15
|
Bal-Price A, Hogberg HT, Crofton KM, Daneshian M, FitzGerald RE, Fritsche E, Heinonen T, Hougaard Bennekou S, Klima S, Piersma AH, Sachana M, Shafer TJ, Terron A, Monnet-Tschudi F, Viviani B, Waldmann T, Westerink RHS, Wilks MF, Witters H, Zurich MG, Leist M. Recommendation on test readiness criteria for new approach methods in toxicology: Exemplified for developmental neurotoxicity. ALTEX-ALTERNATIVES TO ANIMAL EXPERIMENTATION 2018; 35:306-352. [PMID: 29485663 DOI: 10.14573/altex.1712081] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 01/29/2018] [Indexed: 01/06/2023]
Abstract
Multiple non-animal-based test methods have never been formally validated. In order to use such new approach methods (NAMs) in a regulatory context, criteria to define their readiness are necessary. The field of developmental neurotoxicity (DNT) testing is used to exemplify the application of readiness criteria. The costs and number of untested chemicals are overwhelming for in vivo DNT testing. Thus, there is a need for inexpensive, high-throughput NAMs, to obtain initial information on potential hazards, and to allow prioritization for further testing. A background on the regulatory and scientific status of DNT testing is provided showing different types of test readiness levels, depending on the intended use of data from NAMs. Readiness criteria, compiled during a stakeholder workshop, uniting scientists from academia, industry and regulatory authorities are presented. An important step beyond the listing of criteria, was the suggestion for a preliminary scoring scheme. On this basis a (semi)-quantitative analysis process was assembled on test readiness of 17 NAMs with respect to various uses (e.g. prioritization/screening, risk assessment). The scoring results suggest that several assays are currently at high readiness levels. Therefore, suggestions are made on how DNT NAMs may be assembled into an integrated approach to testing and assessment (IATA). In parallel, the testing state in these assays was compiled for more than 1000 compounds. Finally, a vision is presented on how further NAM development may be guided by knowledge of signaling pathways necessary for brain development, DNT pathophysiology, and relevant adverse outcome pathways (AOP).
Collapse
Affiliation(s)
- Anna Bal-Price
- European Commission, Joint Research Centre (EC JRC), Ispra (VA), Italy
| | - Helena T Hogberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Baltimore, MD, USA
| | - Kevin M Crofton
- National Centre for Computational Toxicology, US EPA, RTP, Washington, NC, USA
| | - Mardas Daneshian
- Center for Alternatives to Animal Testing, CAAT-Europe, University of Konstanz, Konstanz, Germany
| | - Rex E FitzGerald
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland
| | - Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine & Heinrich-Heine-University, Düsseldorf, Germany
| | - Tuula Heinonen
- Finnish Centre for Alternative Methods (FICAM), University of Tampere, Tampere, Finland
| | | | - Stefanie Klima
- In vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Aldert H Piersma
- RIVM, National Institute for Public Health and the Environment, Bilthoven, and Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Magdalini Sachana
- Organisation for Economic Co-operation and Development (OECD), Paris, France
| | - Timothy J Shafer
- National Centre for Computational Toxicology, US EPA, RTP, Washington, NC, USA
| | | | - Florianne Monnet-Tschudi
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland.,Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Barbara Viviani
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Tanja Waldmann
- In vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Remco H S Westerink
- Neurotoxicology Research Group, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Martin F Wilks
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland
| | - Hilda Witters
- VITO, Flemish Institute for Technological Research, Unit Environmental Risk and Health, Mol, Belgium
| | - Marie-Gabrielle Zurich
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland.,Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Marcel Leist
- Center for Alternatives to Animal Testing, CAAT-Europe, University of Konstanz, Konstanz, Germany.,In vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Konstanz, Germany
| |
Collapse
|
16
|
Gkioka E, Korou LM, Daskalopoulou A, Misitzi A, Batsidis E, Bakoyiannis I, Pergialiotis V. Prenatal cocaine exposure and its impact on cognitive functions of offspring: a pathophysiological insight. Rev Neurosci 2018; 27:523-34. [PMID: 26953708 DOI: 10.1515/revneuro-2015-0064] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/07/2016] [Indexed: 11/15/2022]
Abstract
It is estimated that approximately 0.5%-3% of fetuses are prenatally exposed to cocaine (COC). The neurodevelopmental implications of this exposure are numerous and include motor skill impairments, alterations of social function, predisposition to anxiety, and memory function and attention deficits; these implications are commonly observed in experimental studies and ultimately affect both learning and IQ. According to previous studies, the clinical manifestations of prenatal COC exposure seem to persist at least until adolescence. The pathophysiological cellular processes that underlie these impairments include dysfunctional myelination, disrupted dendritic architecture, and synaptic alterations. On a molecular level, various neurotransmitters such as serotonin, dopamine, catecholamines, and γ-aminobutyric acid seem to participate in this process. Finally, prenatal COC abuse has been also associated with functional changes in the hormones of the hypothalamic-pituitary-adrenal axis that mediate neuroendocrine responses. The purpose of this review is to summarize the neurodevelopmental consequences of prenatal COC abuse, to describe the pathophysiological pathways that underlie these consequences, and to provide implications for future research in the field.
Collapse
|
17
|
Scott LL, Downing TG. A Single Neonatal Exposure to BMAA in a Rat Model Produces Neuropathology Consistent with Neurodegenerative Diseases. Toxins (Basel) 2017; 10:E22. [PMID: 29286334 PMCID: PMC5793109 DOI: 10.3390/toxins10010022] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 12/23/2017] [Accepted: 12/27/2017] [Indexed: 12/11/2022] Open
Abstract
Although cyanobacterial β-N-methylamino-l-alanine (BMAA) has been implicated in the development of Alzheimer's Disease (AD), Parkinson's Disease (PD) and Amyotrophic Lateral Sclerosis (ALS), no BMAA animal model has reproduced all the neuropathology typically associated with these neurodegenerative diseases. We present here a neonatal BMAA model that causes β-amyloid deposition, neurofibrillary tangles of hyper-phosphorylated tau, TDP-43 inclusions, Lewy bodies, microbleeds and microgliosis as well as severe neuronal loss in the hippocampus, striatum, substantia nigra pars compacta, and ventral horn of the spinal cord in rats following a single BMAA exposure. We also report here that BMAA exposure on particularly PND3, but also PND4 and 5, the critical period of neurogenesis in the rodent brain, is substantially more toxic than exposure to BMAA on G14, PND6, 7 and 10 which suggests that BMAA could potentially interfere with neonatal neurogenesis in rats. The observed selective toxicity of BMAA during neurogenesis and, in particular, the observed pattern of neuronal loss observed in BMAA-exposed rats suggest that BMAA elicits its effect by altering dopamine and/or serotonin signaling in rats.
Collapse
Affiliation(s)
- Laura Louise Scott
- Department of Biochemistry and Microbiology, Nelson Mandela University, P.O. Box 77 000, Port Elizabeth 6031, South Africa.
| | - Timothy Grant Downing
- Department of Biochemistry and Microbiology, Nelson Mandela University, P.O. Box 77 000, Port Elizabeth 6031, South Africa.
| |
Collapse
|
18
|
Scott LL, Downing TG. Β-N-Methylamino-L-Alanine (BMAA) Toxicity Is Gender and Exposure-Age Dependent in Rats. Toxins (Basel) 2017; 10:E16. [PMID: 29280981 PMCID: PMC5793103 DOI: 10.3390/toxins10010016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 12/21/2017] [Accepted: 12/26/2017] [Indexed: 12/21/2022] Open
Abstract
Cyanobacterial β-N-methylamino-L-alanine (BMAA) has been suggested as a causative or contributory factor in the development of several neurodegenerative diseases. However, no BMAA animal model has adequately shown clinical or behavioral symptoms that correspond to those seen in either Alzheimer's Disease (AD), Amyotrophic Lateral Sclerosis (ALS) or Parkinson's Disease (PD). We present here the first data that show that when neonatal rats were exposed to BMAA on postnatal days 3, 4 and 5, but not on gestational day 14 or postnatally on days 7 or 10, several AD and/or PD-related behavioral, locomotor and cognitive deficits developed. Male rats exhibited severe unilateral hindlimb splay while whole body tremors could be observed in exposed female rats. BMAA-exposed rats failed to identify and discriminate a learned odor, an early non-motor symptom of PD, and exhibited decreased locomotor activity, decreased exploration and increased anxiety in the open field test. Alterations were also observed in the rats' natural passive defense mechanism, and potential memory deficits and changes to the rat's natural height avoidance behavior could be observed as early as PND 30. Spatial learning, short-term working, reference and long-term memory were also impaired in 90-day-old rats that had been exposed to a single dose of BMAA on PND 3-7. These data suggest that BMAA is a developmental neurotoxin, with specific target areas in the brain and spinal cord.
Collapse
Affiliation(s)
- Laura Louise Scott
- Department of Biochemistry and Microbiology, Nelson Mandela University, P.O. Box 77 000, Port Elizabeth 6031, South Africa.
| | - Timothy Grant Downing
- Department of Biochemistry and Microbiology, Nelson Mandela University, P.O. Box 77 000, Port Elizabeth 6031, South Africa.
| |
Collapse
|
19
|
Obi K, Amano I, Takatsuru Y. Role of dopamine on functional recovery in the contralateral hemisphere after focal stroke in the somatosensory cortex. Brain Res 2017; 1678:146-152. [PMID: 29079503 DOI: 10.1016/j.brainres.2017.10.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/03/2017] [Accepted: 10/22/2017] [Indexed: 01/05/2023]
Abstract
Functional recovery after a stroke is important for patients' quality of life. Not only medical care during the acute phase, but also rehabilitation during the chronic phase after a stroke is important. However, the mechanisms underlying functional recovery, particularly the chronic phase after stroke, are still not fully understood. Thus, further basic study on brain after focal stroke is necessary. In this study, we found that the concentration of dopamine (DA) increased during first week after a stroke in the hemisphere contralateral in the site of stroke by in vivo microdialysis. When we applied haloperidol (HPD), a potent DA receptor blocker, functional recovery was inhibited. Interestingly, administration of aripiprazole (ARP), a novel partial agonist of the DA receptor, during the chronic phase improved the remodeling of neuronal circuits in somatosensory cortex (SSC). These findings indicate that the DAergic system play a critical role in functional compensation by the non-infarcted hemisphere after a focal stroke in SSC. It is also revealed that administration of HPD/ARP to stroke patients affects functional recovery after a stroke, and stimulation of the DAergic system during the chronic phase of stroke potentially benefits stroke patients.
Collapse
Affiliation(s)
- Kisho Obi
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Izuki Amano
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Yusuke Takatsuru
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan.
| |
Collapse
|
20
|
Abstract
BACKGROUND A previous survey of the literature of fMRI brain activation for two risk factors, impulsivity and craving, for addiction were lateralized to the right and left hemispheres respectively. Most articles reported these findings without consideration of how lateral asymmetries might be relevant to understanding the underlying factors leading to addiction. OBJECTIVE The current survey is intended to extend these observations by demonstrating hemispheric asymmetry of development due to pre-natal or adolescent/adult exposure to drugs of abuse. METHOD Articles that reported either pre-natal or adolescent/adult exposure to drugs of abuse were collected and the hemisphere of the affected structures was tabulated to determine if, and which, drugs affected more structures in one hemisphere or the other or both together. RESULTS Some drugs, notably cocaine and alcohol, differentially affected left or right hemisphere structures which significantly differed depending on whether individuals were exposed prenatally or as an adolescent/adult. Cocaine tended to affect more left hemisphere structures when exposed prenatally and significantly affected more in the right when exposed as adults. Alcohol had the reverse pattern. The difference in patterns of effect between pre-natal or adult exposure was significant for both. CONCLUSION The results in this survey demonstrate that some drugs of abuse appear to have a right/left differential effect on structures of the brain. Further investigation into the reasons for this asymmetry may provide new insights into underlying factors of drug-seeking and addiction.
Collapse
Affiliation(s)
- Harold W Gordon
- Epidemiology Research Branch, Division of Epidemiology, Services, and Prevention Research, National Institute on Drug Abuse
| |
Collapse
|
21
|
Azzarelli R, Oleari R, Lettieri A, Andre' V, Cariboni A. In Vitro, Ex Vivo and In Vivo Techniques to Study Neuronal Migration in the Developing Cerebral Cortex. Brain Sci 2017; 7:brainsci7050048. [PMID: 28448448 PMCID: PMC5447930 DOI: 10.3390/brainsci7050048] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 04/21/2017] [Accepted: 04/24/2017] [Indexed: 11/16/2022] Open
Abstract
Neuronal migration is a fundamental biological process that underlies proper brain development and neuronal circuit formation. In the developing cerebral cortex, distinct neuronal populations, producing excitatory, inhibitory and modulatory neurotransmitters, are generated in different germinative areas and migrate along various routes to reach their final positions within the cortex. Different technical approaches and experimental models have been adopted to study the mechanisms regulating neuronal migration in the cortex. In this review, we will discuss the most common in vitro, ex vivo and in vivo techniques to visualize and study cortical neuronal migration.
Collapse
Affiliation(s)
- Roberta Azzarelli
- Department of Oncology, University of Cambridge, Hutchison-MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK.
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK.
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge CB3 0HE, UK.
| | - Roberto Oleari
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9, Milan 20133, Italy.
| | - Antonella Lettieri
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9, Milan 20133, Italy.
| | - Valentina Andre'
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9, Milan 20133, Italy.
| | - Anna Cariboni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9, Milan 20133, Italy.
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK.
| |
Collapse
|
22
|
Mardini V, Rohde LA, Ceresér KM, Gubert CM, Silva EGD, Xavier F, Parcianello R, Röhsig LM, Pechansky F, Szobot CM. TBARS and BDNF levels in newborns exposed to crack/cocaine during pregnancy: a comparative study. ACTA ACUST UNITED AC 2017; 39:263-266. [PMID: 28273279 PMCID: PMC7111386 DOI: 10.1590/1516-4446-2016-2035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/28/2016] [Indexed: 12/31/2022]
Abstract
Objectives: To compare levels of a marker of lipid peroxidation (thiobarbituric acid reactive substances, TBARS) and brain-derived neurotrophic factor (BDNF) in umbilical cord blood (UCB) between newborns exposed to crack/cocaine in utero (exposed newborns [EN], n=57) and non-exposed newborns (NEN, n=99), as well as in maternal peripheral blood at delivery. Methods: This was a cross-sectional study. Potential confounders, including perinatal parameters, psychopathology, and use of other substances, were assessed. Results: After adjusting for potential confounders, adjusted mean BDNF was significantly higher in EN (3.86 ng/mL, 95% confidence interval [95%CI] 2.29-5.43) than in NEN (0.85 ng/mL, 95%CI 0.47-1.23; p < 0.001; Cohen effect size: 1.12), and significantly lower in crack/cocaine mothers than in control mothers (4.03 ng/mL, 95%CI 2.87-5.18 vs. 6.67 ng/mL, 95%CI 5.60-7.74; p = 0.006). The adjusted mean TBARS level was significantly lower in EN (63.97 µM MDA, 95%CI 39.43-88.50) than NEN (177.04 µM MDA, 95%CI 140.93-213.14; p < 0.001; effect size = 0.84), with no difference between mother groups (p = 0.86). Conclusions: The changes in TBARS levels observed in EN suggest that fetuses exposed to cocaine mobilize endogenous antioxidant routes since very early stages of development. The increase in BDNF levels in EN might indicate changes in fetal development, whereas the changes in BDNF levels in mothers provide evidence of the complex metabolic processes involved in drug use during pregnancy.
Collapse
Affiliation(s)
- Victor Mardini
- Serviço de Psiquiatria da Infância e Adolescência (SPIA), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Medicina: Psiquiatria, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Luis A Rohde
- Serviço de Psiquiatria da Infância e Adolescência (SPIA), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Medicina: Psiquiatria, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.,Instituto Nacional de Psiquiatria do Desenvolvimento (INPD), São Paulo, SP, Brazil.,Departamento de Psiquiatria, UFRGS, Porto Alegre, RS, Brazil
| | - Keila M Ceresér
- Programa de Pós-Graduação em Medicina: Psiquiatria, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.,Laboratório de Psiquiatria Molecular, Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), HCPA, UFRGS, Porto Alegre, RS, Brazil
| | - Carolina M Gubert
- Laboratório de Psiquiatria Molecular, Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), HCPA, UFRGS, Porto Alegre, RS, Brazil
| | - Emily G da Silva
- Laboratório de Psiquiatria Molecular, Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), HCPA, UFRGS, Porto Alegre, RS, Brazil
| | - Fernando Xavier
- Faculdade de Ciências Biomédicas, Centro Universitário Metodista IPA, Porto Alegre, RS, Brazil
| | - Rodrigo Parcianello
- Programa de Pós-Graduação em Medicina: Psiquiatria, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Liane M Röhsig
- Unidade de Criobiologia, Banco de Cordão Umbilical e Placentário, HCPA, Porto Alegre, RS, Brazil
| | - Flávio Pechansky
- Programa de Pós-Graduação em Medicina: Psiquiatria, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.,Departamento de Psiquiatria, UFRGS, Porto Alegre, RS, Brazil.,Centro de Pesquisa em Álcool e Drogas, HCPA, UFRGS, Porto Alegre, RS, Brazil
| | - Claudia M Szobot
- Serviço de Psiquiatria da Infância e Adolescência (SPIA), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Medicina: Psiquiatria, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.,Centro de Pesquisa em Álcool e Drogas, HCPA, UFRGS, Porto Alegre, RS, Brazil
| |
Collapse
|
23
|
McCarthy DM, Bell GA, Cannon EN, Mueller KA, Huizenga MN, Sadri-Vakili G, Fadool DA, Bhide PG. Reversal Learning Deficits Associated with Increased Frontal Cortical Brain-Derived Neurotrophic Factor Tyrosine Kinase B Signaling in a Prenatal Cocaine Exposure Mouse Model. Dev Neurosci 2016; 38:354-364. [PMID: 27951531 PMCID: PMC5360472 DOI: 10.1159/000452739] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 10/20/2016] [Indexed: 12/20/2022] Open
Abstract
Prenatal cocaine exposure remains a major public health concern because of its adverse impact on cognitive function in children and adults. We report that prenatal cocaine exposure produces significant deficits in reversal learning, a key component of cognitive flexibility, in a mouse model. We used an olfactory reversal learning paradigm and found that the prenatally cocaine-exposed mice showed a marked failure to learn the reversed paradigm. Because brain-derived neurotrophic factor (BDNF) is a key regulator of cognitive functions, and because prenatal cocaine exposure increases the expression of BDNF and the phosphorylated form of its receptor, tyrosine kinase B (TrkB), we examined whether BDNF-TrkB signaling is involved in mediating the reversal learning deficit in prenatally cocaine-exposed mice. Systemic administration of a selective TrkB receptor antagonist restored normal reversal learning in prenatally cocaine-exposed mice, suggesting that increased BDNF-TrkB signaling may be an underlying mechanism of reversal learning deficits. Our findings provide novel mechanistic insights into the reversal learning phenomenon and may have significant translational implications because impaired cognitive flexibility is a key symptom in psychiatric conditions of developmental onset.
Collapse
Affiliation(s)
- Deirdre M. McCarthy
- Center for Brain Repair, Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, 32306
| | - Genevieve A. Bell
- Department of Biological Science, Florida State University, Tallahassee, FL, 32306
- Program in Neuroscience, Florida State University, Tallahassee, FL, 32306
| | - Elisa N. Cannon
- Center for Brain Repair, Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, 32306
| | - Kaly A. Mueller
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129-4404
| | - Megan N. Huizenga
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129-4404
| | - Ghazaleh Sadri-Vakili
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129-4404
| | - Debra A. Fadool
- Department of Biological Science, Florida State University, Tallahassee, FL, 32306
- Program in Neuroscience, Florida State University, Tallahassee, FL, 32306
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, 32306
| | - Pradeep G. Bhide
- Center for Brain Repair, Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, 32306
- Program in Neuroscience, Florida State University, Tallahassee, FL, 32306
| |
Collapse
|
24
|
Abstract
T-box transcription factors play key roles in the regulation of developmental processes such as cell differentiation and migration. Mammals have 17 T-box genes, of which several regulate brain development. The Tbr1 subfamily of T-box genes is particularly important in development of the cerebral cortex, olfactory bulbs (OBs), and cerebellum. This subfamily is comprised of Tbr1, Tbr2 (also known as Eomes), and Tbx21. In developing cerebral cortex, Tbr2 and Tbr1 are expressed during successive stages of differentiation in the pyramidal neuron lineage, from Tbr2+ intermediate progenitors to Tbr1+ postmitotic glutamatergic neurons. At each stage, Tbr2 and Tbr1 regulate laminar and regional identity of cortical projection neurons, cell migration, and axon guidance. In the OB, Tbr1 subfamily genes regulate neurogenesis of mitral and tufted cells, and glutamatergic juxtaglomerular interneurons. Tbr2 is also prominent in the development of retinal ganglion cells in nonimage-forming pathways. Other regions that require Tbr2 or Tbr1 in development or adulthood include the cerebellum and adult dentate gyrus. In humans, de novo mutations in TBR1 are important causes of sporadic autism and intellectual disability. Further studies of T-box transcription factors will enhance our understanding of neurodevelopmental disorders and inform approaches to new therapies.
Collapse
|
25
|
Stucky A, Bakshi KP, Friedman E, Wang HY. Prenatal Cocaine Exposure Upregulates BDNF-TrkB Signaling. PLoS One 2016; 11:e0160585. [PMID: 27494324 PMCID: PMC4975466 DOI: 10.1371/journal.pone.0160585] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 07/21/2016] [Indexed: 12/11/2022] Open
Abstract
Prenatal cocaine exposure causes profound changes in neurobehavior as well as synaptic function and structure with compromised glutamatergic transmission. Since synaptic health and glutamatergic activity are tightly regulated by brain-derived neurotrophic factor (BDNF) signaling through its cognate tyrosine receptor kinase B (TrkB), we hypothesized that prenatal cocaine exposure alters BDNF-TrkB signaling during brain development. Here we show prenatal cocaine exposure enhances BDNF-TrkB signaling in hippocampus and prefrontal cortex (PFCX) of 21-day-old rats without affecting the expression levels of TrkB, P75NTR, signaling molecules, NMDA receptor—NR1 subunit as well as proBDNF and BDNF. Prenatal cocaine exposure reduces activity-dependent proBDNF and BDNF release and elevates BDNF affinity for TrkB leading to increased tyrosine-phosphorylated TrkB, heightened Phospholipase C-γ1 and N-Shc/Shc recruitment and higher downstream PI3K and ERK activation in response to ex vivo BDNF. The augmented BDNF-TrkB signaling is accompanied by increases in association between activated TrkB and NMDARs. These data suggest that cocaine exposure during gestation upregulates BDNF-TrkB signaling and its interaction with NMDARs by increasing BDNF affinity, perhaps in an attempt to restore the diminished excitatory neurotransmission.
Collapse
Affiliation(s)
- Andres Stucky
- Departments of Physiology, Pharmacology and Neuroscience, School of Medicine at CCNY, The City University of New York, New York, New York, 10031, United States of America
- Department of Biology, Neuroscience Program, Graduate School of The City University of New York, New York, New York, 10061, United States of America
| | - Kalindi P. Bakshi
- Departments of Physiology, Pharmacology and Neuroscience, School of Medicine at CCNY, The City University of New York, New York, New York, 10031, United States of America
| | - Eitan Friedman
- Departments of Physiology, Pharmacology and Neuroscience, School of Medicine at CCNY, The City University of New York, New York, New York, 10031, United States of America
| | - Hoau-Yan Wang
- Departments of Physiology, Pharmacology and Neuroscience, School of Medicine at CCNY, The City University of New York, New York, New York, 10031, United States of America
- * E-mail:
| |
Collapse
|
26
|
Scott-Goodwin A, Puerto M, Moreno I. Toxic effects of prenatal exposure to alcohol, tobacco and other drugs. Reprod Toxicol 2016; 61:120-30. [DOI: 10.1016/j.reprotox.2016.03.043] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 03/08/2016] [Accepted: 03/28/2016] [Indexed: 01/21/2023]
|
27
|
Martin MM, Graham DL, McCarthy DM, Bhide PG, Stanwood GD. Cocaine-induced neurodevelopmental deficits and underlying mechanisms. BIRTH DEFECTS RESEARCH. PART C, EMBRYO TODAY : REVIEWS 2016; 108:147-73. [PMID: 27345015 PMCID: PMC5538582 DOI: 10.1002/bdrc.21132] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 06/06/2016] [Indexed: 11/06/2022]
Abstract
Exposure to drugs early in life has complex and long-lasting implications for brain structure and function. This review summarizes work to date on the immediate and long-term effects of prenatal exposure to cocaine. In utero cocaine exposure produces disruptions in brain monoamines, particularly dopamine, during sensitive periods of brain development, and leads to permanent changes in specific brain circuits, molecules, and behavior. Here, we integrate clinical studies and significance with mechanistic preclinical studies, to define our current knowledge base and identify gaps for future investigation. Birth Defects Research (Part C) 108:147-173, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Melissa M. Martin
- Department of Biomedical Sciences and Center for Brain Repair, Florida State University College of Medicine, Tallahassee, Florida
| | - Devon L. Graham
- Department of Biomedical Sciences and Center for Brain Repair, Florida State University College of Medicine, Tallahassee, Florida
| | - Deirdre M. McCarthy
- Department of Biomedical Sciences and Center for Brain Repair, Florida State University College of Medicine, Tallahassee, Florida
| | - Pradeep G. Bhide
- Department of Biomedical Sciences and Center for Brain Repair, Florida State University College of Medicine, Tallahassee, Florida
| | - Gregg D. Stanwood
- Department of Biomedical Sciences and Center for Brain Repair, Florida State University College of Medicine, Tallahassee, Florida
| |
Collapse
|
28
|
Trentini JF, O'Neill JT, Poluch S, Juliano SL. Prenatal carbon monoxide impairs migration of interneurons into the cerebral cortex. Neurotoxicology 2015; 53:31-44. [PMID: 26582457 DOI: 10.1016/j.neuro.2015.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 11/09/2015] [Accepted: 11/09/2015] [Indexed: 01/15/2023]
Abstract
Prenatal exposure to carbon monoxide (CO) disrupts brain development, however little is known about effects on neocortical maturation. We exposed pregnant mice to CO from embryonic day 7 (E7) until birth. To study the effect of CO on neuronal migration into the neocortex we injected BrdU during corticogenesis and observed misplaced BrdU+ cells. The majority of cells not in their proper layer colocalized with GAD65/67, suggesting impairment of interneuron migration; interneuron subtypes were also affected. We subsequently followed interneuron migration from E15 organotypic cultures of mouse neocortex exposed to CO; the leading process length of migrating neurons diminished. To examine an underlying mechanism, we assessed the effects of CO on the cellular cascade mediating the cytoskeletal protein vasodilator-stimulated phosphoprotein (VASP). CO exposure resulted in decreased cGMP and in a downstream target, phosphorylated VASP. Organotypic cultures grown in the presence of the phosphodiesterase inhibitor IBMX resulted in a recovery of the leading processes. These data support the idea that CO acts as a signaling molecule and impairs function and neuronal migration by acting through the CO/NO-cGMP pathway. In addition, treated mice demonstrated functional impairment in behavioral tests.
Collapse
Affiliation(s)
- John F Trentini
- Graduate Program in Neuroscience, Uniformed Services University, Bethesda, MD 20814, USA
| | - J Timothy O'Neill
- Graduate Program in Neuroscience, Uniformed Services University, Bethesda, MD 20814, USA; Department of Pediatrics, Uniformed Services University, Bethesda, MD 20814, USA
| | - Sylvie Poluch
- Graduate Program in Neuroscience, Uniformed Services University, Bethesda, MD 20814, USA; Department of Anatomy, Physiology and Genetics, Uniformed Services University, Bethesda, MD 20814, USA
| | - Sharon L Juliano
- Graduate Program in Neuroscience, Uniformed Services University, Bethesda, MD 20814, USA; Department of Anatomy, Physiology and Genetics, Uniformed Services University, Bethesda, MD 20814, USA.
| |
Collapse
|
29
|
Pereira RB, Andrade PB, Valentão P. A Comprehensive View of the Neurotoxicity Mechanisms of Cocaine and Ethanol. Neurotox Res 2015; 28:253-67. [PMID: 26105693 DOI: 10.1007/s12640-015-9536-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/09/2015] [Accepted: 06/16/2015] [Indexed: 01/17/2023]
Abstract
Substance use disorder is an emerging problem concerning to human health, causing severe side effects, including neurotoxicity. The use of illegal drugs and the misuse of prescription or over-the-counter drugs are growing in this century, being one of the major public health problems. Ethanol and cocaine are one of the most frequently used drugs and, according to the National Institute on Drug Abuse, their concurrent consumption is one of the major causes for emergency hospital room visits. These molecules act in the brain through different mechanisms, altering the nervous system function. Researchers have focused the attention not just in the mechanism of action of these drugs, but also in the mechanism by which they damage the nervous tissue (neurotoxicity). Therefore, the goal of the present review is to provide a global perspective about the mechanisms of the neurotoxicity of cocaine and ethanol.
Collapse
Affiliation(s)
- Renato B Pereira
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, nº 228, 4050-313, Porto, Portugal
| | | | | |
Collapse
|
30
|
Zhang L, McCarthy DM, Sharma N, Bhide PG. Dopamine receptor and Gα(olf) expression in DYT1 dystonia mouse models during postnatal development. PLoS One 2015; 10:e0123104. [PMID: 25860259 PMCID: PMC4393110 DOI: 10.1371/journal.pone.0123104] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 02/27/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND DYT1 dystonia is a heritable, early-onset generalized movement disorder caused by a GAG deletion (ΔGAG) in the DYT1 gene. Neuroimaging studies and studies using mouse models suggest that DYT1 dystonia is associated with dopamine imbalance. However, whether dopamine imbalance is key to DYT1 or other forms of dystonia continues to be debated. METHODOLOGY/PRINCIPAL FINDINGS We used Dyt1 knock out (Dyt1 KO), Dyt1 ΔGAG knock-in (Dyt1 KI), and transgenic mice carrying one copy of the human DYT1 wild type allele (DYT1 hWT) or human ΔGAG mutant allele (DYT1 hMT). D1R, D2R, and Gα(olf) protein expression was analyzed by western blot in the frontal cortex, caudate-putamen and ventral midbrain in young adult (postnatal day 60; P60) male mice from all four lines; and in the frontal cortex and caudate putamen in juvenile (postnatal day 14; P14) male mice from the Dyt1 KI and KO lines. Dopamine receptor and Gα(olf) protein expression were significantly decreased in multiple brain regions of Dyt1 KI and Dyt1 KO mice and not significantly altered in the DYT1 hMT or DYT1 hWT mice at P60. The only significant change at P14 was a decrease in D1R expression in the caudate-putamen of the Dyt1 KO mice. CONCLUSION/SIGNIFICANCE We found significant decreases in key proteins in the dopaminergic system in multiple brain regions of Dyt1 KO and Dyt1 KI mouse lines at P60. Deletion of one copy of the Dyt1 gene (KO mice) produced the most pronounced effects. These data offer evidence that impaired dopamine receptor signaling may be an early and significant contributor to DYT1 dystonia pathophysiology.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Biomedical Sciences, Center for Brain Repair, Florida State University College of Medicine, Tallahassee, Florida, United States of America
- * E-mail: (LZ); (PGB)
| | - Deirdre M. McCarthy
- Department of Biomedical Sciences, Center for Brain Repair, Florida State University College of Medicine, Tallahassee, Florida, United States of America
| | - Nutan Sharma
- Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Pradeep G. Bhide
- Department of Biomedical Sciences, Center for Brain Repair, Florida State University College of Medicine, Tallahassee, Florida, United States of America
- * E-mail: (LZ); (PGB)
| |
Collapse
|
31
|
Graham DL, Durai HH, Garden JD, Cohen EL, Echevarria FD, Stanwood GD. Loss of dopamine D2 receptors increases parvalbumin-positive interneurons in the anterior cingulate cortex. ACS Chem Neurosci 2015; 6:297-305. [PMID: 25393953 PMCID: PMC4372074 DOI: 10.1021/cn500235m] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
![]()
Disruption
to dopamine homeostasis during brain development has been implicated
in a variety of neuropsychiatric disorders, including depression and
schizophrenia. Inappropriate expression or activity of GABAergic interneurons
are common features of many of these disorders. We discovered a persistent
upregulation of GAD67+ and parvalbumin+ neurons within the anterior
cingulate cortex of dopamine D2 receptor knockout mice, while other
GABAergic interneuron markers were unaffected. Interneuron distribution
and number were not altered in the striatum or in the dopamine-poor
somatosensory cortex. The changes were already present by postnatal
day 14, indicating a developmental etiology. D2eGFP BAC transgenic
mice demonstrated the presence of D2 receptor expression within a
subset of parvalbumin-expressing cortical interneurons, suggesting
the possibility of a direct cellular mechanism through which D2 receptor
stimulation regulates interneuron differentiation or survival. D2
receptor knockout mice also exhibited decreased depressive-like behavior
compared with wild-type controls in the tail suspension test. These
data indicate that dopamine signaling modulates interneuron number
and emotional behavior and that developmental D2 receptor loss or
blockade could reveal a potential mechanism for the prodromal basis
of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Devon L. Graham
- Department of Pharmacology, ‡Vanderbilt Brain Institute, §Vanderbilt Kennedy
Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Heather H. Durai
- Department of Pharmacology, ‡Vanderbilt Brain Institute, §Vanderbilt Kennedy
Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Jamie D. Garden
- Department of Pharmacology, ‡Vanderbilt Brain Institute, §Vanderbilt Kennedy
Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Evan L. Cohen
- Department of Pharmacology, ‡Vanderbilt Brain Institute, §Vanderbilt Kennedy
Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Franklin D. Echevarria
- Department of Pharmacology, ‡Vanderbilt Brain Institute, §Vanderbilt Kennedy
Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Gregg D. Stanwood
- Department of Pharmacology, ‡Vanderbilt Brain Institute, §Vanderbilt Kennedy
Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
32
|
Nery FC, da Hora CC, Yaqub U, Zhang X, McCarthy DM, Bhide PG, Irimia D, Breakefield XO. New methods for investigation of neuronal migration in embryonic brain explants. J Neurosci Methods 2015; 239:80-4. [PMID: 25291524 PMCID: PMC4268085 DOI: 10.1016/j.jneumeth.2014.09.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 09/24/2014] [Accepted: 09/25/2014] [Indexed: 01/19/2023]
Abstract
BACKGROUND Proper migration of neurons is essential for the formation and normal functioning of the nervous system. Defects in neuronal migration underlie a number of neurologic diseases in humans. Although cell migration is crucial for neural development, molecular mechanisms guiding neuronal migration remain to be elucidated fully. Newborn neurons from the embryonic medial ganglionic eminence (MGE) migrate a long distance dorsally in the developing brain, giving rise to several types of interneurons in the neocortex. NEW METHOD In this study, we developed an immunocytochemistry (ICC) protocol to stain neurons migrating out of the MGE explant embedded in Matrigel. We also established a protocol to efficiently transfect cells in MGE explants, achieving a transduction efficiency of more than 30%. COMPARISON WITH EXISTING METHOD In addition, we developed microfluidic chambers for explants that allow visualization of the vectorial migration of individual neurons from mouse embryonic MGE explants. Our microfluidic system allows monitoring of the distribution of cellular organelles (e.g. Golgi) within migrating neurons which have been stained with commercial molecular dyes or transfected with adeno-associated virus (AAV) expressing reporter proteins. CONCLUSION These methods provide new paradigms to study neuronal migration in real-time.
Collapse
Affiliation(s)
- Flávia C Nery
- Department of Neurology and Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA 02114, USA.
| | - Cintia C da Hora
- Department of Neurology and Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA 02114, USA
| | - Uzma Yaqub
- Department of Neurology and Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA 02114, USA
| | - Xuan Zhang
- Department of Neurology and Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA 02114, USA
| | - Deirdre M McCarthy
- Department of Neurology and Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA 02114, USA; Center for Brain Repair, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Pradeep G Bhide
- Department of Neurology and Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA 02114, USA; Center for Brain Repair, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Daniel Irimia
- BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Charlestown, MA 02129 USA
| | - Xandra O Breakefield
- Department of Neurology and Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
33
|
Sasaki A, Constantinof A, Pan P, Kupferschmidt DA, McGowan PO, Erb S. Cocaine exposure prior to pregnancy alters the psychomotor response to cocaine and transcriptional regulation of the dopamine D1 receptor in adult male offspring. Behav Brain Res 2014; 265:163-70. [PMID: 24583058 DOI: 10.1016/j.bbr.2014.02.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 01/23/2014] [Accepted: 02/13/2014] [Indexed: 02/01/2023]
Abstract
There is evidence that maternal experience prior to pregnancy can play an important role in behavioral, physiological, and genetic programming of offspring. Likewise, exposure to cocaine in utero can result in marked changes in central nervous system function of offspring. In this study, we examined whether exposure of rat dams to cocaine prior to pregnancy subsequently alters indices of behavior, physiology, and gene expression in offspring. Multiple outcome measures were examined in adult male offspring: (1) behavioral expression of cocaine-induced psychomotor activation; (2) levels of corticosterone in response to immobilization stress; and (3) expression of multiple genes, including dopamine receptor D1 (DRD1) and D2 (DRD2), glucocorticoid receptor (GR), and corticotropin-releasing factor (CRF), in functionally relevant brain regions. Adult Sprague-Dawley females were exposed to cocaine (15-30 mg/kg, i.p.) or saline for 10 days, and were then mated to drug naïve males of the same strain. Separate groups of adult male offspring were tested for their acute psychomotor response to cocaine (0, 15, 30 mg/kg, i.p.), corticosterone responsivity to 20 min of immobilization stress, and expression of multiple genes using quantitative PCR. Offspring of dams exposed to cocaine prior to conception exhibited increased psychomotor sensitivity to cocaine, and upregulated gene expression of DRD1 in the medial prefrontal cortex (mPFC). Neither stress-induced corticosterone levels nor gene expression of GR or CRF genes were altered. These data suggest that cocaine exposure before pregnancy can serve to enhance psychomotor sensitivity to cocaine in offspring, possibly via alterations in dopamine function that include upregulation of the DRD1.
Collapse
Affiliation(s)
- Aya Sasaki
- Centre for the Neurobiology of Stress, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4, Canada; Department of Cell and Systems Biology, University of Toronto, 1265 Military Trail, Toronto, ON M1C 1A4, Canada
| | - Andrea Constantinof
- Department of Psychology, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4, Canada
| | - Pauline Pan
- Department of Cell and Systems Biology, University of Toronto, 1265 Military Trail, Toronto, ON M1C 1A4, Canada
| | - Dave A Kupferschmidt
- Centre for the Neurobiology of Stress, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4, Canada; Department of Psychology, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4, Canada
| | - Patrick O McGowan
- Centre for the Neurobiology of Stress, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4, Canada; Department of Cell and Systems Biology, University of Toronto, 1265 Military Trail, Toronto, ON M1C 1A4, Canada
| | - Suzanne Erb
- Centre for the Neurobiology of Stress, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4, Canada; Department of Psychology, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4, Canada; Department of Cell and Systems Biology, University of Toronto, 1265 Military Trail, Toronto, ON M1C 1A4, Canada.
| |
Collapse
|
34
|
McCarthy DM, Kabir ZD, Bhide PG, Kosofsky BE. Effects of prenatal exposure to cocaine on brain structure and function. PROGRESS IN BRAIN RESEARCH 2014; 211:277-89. [DOI: 10.1016/b978-0-444-63425-2.00012-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
35
|
Money KM, Stanwood GD. Developmental origins of brain disorders: roles for dopamine. Front Cell Neurosci 2013; 7:260. [PMID: 24391541 PMCID: PMC3867667 DOI: 10.3389/fncel.2013.00260] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 12/01/2013] [Indexed: 01/11/2023] Open
Abstract
Neurotransmitters and neuromodulators, such as dopamine, participate in a wide range of behavioral and cognitive functions in the adult brain, including movement, cognition, and reward. Dopamine-mediated signaling plays a fundamental neurodevelopmental role in forebrain differentiation and circuit formation. These developmental effects, such as modulation of neuronal migration and dendritic growth, occur before synaptogenesis and demonstrate novel roles for dopaminergic signaling beyond neuromodulation at the synapse. Pharmacologic and genetic disruptions demonstrate that these effects are brain region- and receptor subtype-specific. For example, the striatum and frontal cortex exhibit abnormal neuronal structure and function following prenatal disruption of dopamine receptor signaling. Alterations in these processes are implicated in the pathophysiology of neuropsychiatric disorders, and emerging studies of neurodevelopmental disruptions may shed light on the pathophysiology of abnormal neuronal circuitry in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Kelli M Money
- Neuroscience Graduate Program, Vanderbilt University Nashville, TN, USA ; Vanderbilt Medical Scientist Training Program, Vanderbilt University Nashville, TN, USA
| | - Gregg D Stanwood
- Department of Pharmacology, Vanderbilt University Nashville, TN, USA ; Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University Nashville, TN, USA
| |
Collapse
|
36
|
Chavez-Valdez R, Martin LJ, Razdan S, Gauda EB, Northington FJ. Sexual dimorphism in BDNF signaling after neonatal hypoxia-ischemia and treatment with necrostatin-1. Neuroscience 2013; 260:106-19. [PMID: 24361177 DOI: 10.1016/j.neuroscience.2013.12.023] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 11/17/2013] [Accepted: 12/10/2013] [Indexed: 12/30/2022]
Abstract
Brain injury due to neonatal hypoxia-ischemia (HI) is more homogenously severe in male than in female mice. Because, necrostatin-1 (nec-1) prevents injury progression only in male mice, we hypothesized that changes in brain-derived neurotrophic factor (BDNF) signaling after HI and nec-1 are also sex-specific providing differential conditions to promote recovery of those more severely injured. The increased aromatization of testosterone in male mice during early development and the link between 17-β-estradiol (E2) levels and BDNF transcription substantiate this hypothesis. Hence, we aimed to investigate if sexual differences in BDNF signaling existed in forebrain and diencephalon after HI and HI/nec-1 and their correlation with estrogen receptors (ER). C57B6 mice (p7) received nec-1 (0.1μl [8μM]) or vehicle (veh) intracerebroventricularly after HI. At 24h after HI, BDNF levels increased in both sexes in forebrain without evidence of tropomyosin-receptor-kinase B (TrkB) activation. At 96h after HI, BDNF levels in forebrain decreased below those seen in control mice of both sexes. Additionally, only in female mice, truncated TrkB (Tc.TrkB) and p75 neurotrophic receptor (p75ntr) levels increased in forebrain and diencephalon. In both, forebrain and diencephalon, nec-1 treatment increased BDNF levels and TrkB activation in male mice while, nec-1 prevented Tc.TrkB and p75ntr increases in female mice. While E2 levels were unchanged by HI or HI/nec-1 in either sex or treatment, ERα:ERβ ratios were increased in diencephalon of nec-1-treated male mice and directly correlated with BDNF levels. Neonatal HI produces sex-specific signaling changes in the BDNF system, that are differentially modulated by nec-1. The regional differences in BDNF levels may be a consequence of injury severity after HI, but sexual differences in response to nec-1 after HI may represent a differential thalamo-cortical preservation or alternatively off-target regional effect of nec-1. The biological significance of ERα predominance and its correlation with BDNF levels is still unclear.
Collapse
Affiliation(s)
- R Chavez-Valdez
- Department of Pediatrics, Neonatal Research Laboratory, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 6-104, Baltimore, MD 21287, USA.
| | - L J Martin
- Department of Pathology, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Research Building, Room 558, Baltimore, MD 21205, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Research Building, Room 558, Baltimore, MD 21205, USA
| | - S Razdan
- Department of Pediatrics, Neonatal Research Laboratory, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 6-104, Baltimore, MD 21287, USA
| | - E B Gauda
- Department of Pediatrics, Neonatal Research Laboratory, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 6-104, Baltimore, MD 21287, USA
| | - F J Northington
- Department of Pediatrics, Neonatal Research Laboratory, Johns Hopkins University School of Medicine, 600 North Wolfe Street, CMSC 6-104, Baltimore, MD 21287, USA
| |
Collapse
|
37
|
Marín O. Cellular and molecular mechanisms controlling the migration of neocortical interneurons. Eur J Neurosci 2013; 38:2019-29. [DOI: 10.1111/ejn.12225] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 03/18/2013] [Accepted: 03/21/2013] [Indexed: 12/16/2022]
Affiliation(s)
- Oscar Marín
- Instituto de Neurociencias; Consejo Superior de Investigaciones Científicas; Universidad Miguel Hernández; Sant Joan d'Alacant; Spain
| |
Collapse
|
38
|
McCarthy DM, Brown AN, Bhide PG. Regulation of BDNF expression by cocaine. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2012; 85:437-46. [PMID: 23239946 PMCID: PMC3516887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin family of growth factors. It is expressed throughout the nervous system. A unique feature of the BDNF gene is the existence of multiple mRNA transcripts, all of which are translated into BDNF protein, suggesting a multilevel regulation of expression. In particular, the BDNF exon IV promoter region is a preferential target for epigenetic alterations, as it contains binding sites for CREB and MeCP2, two transcriptional regulators known to mediate epigenetic changes. Exposure to drugs of abuse is known to modulate epigenetic regulation of BDNF gene expression. This review will discuss how exposure to cocaine, one of the most addictive drugs known to mankind, can produce alterations in BDNF gene expression, especially in the mesolimbic dopaminergic system, which lead to alterations in the reward-mediated behaviors involved in addiction.
Collapse
Affiliation(s)
- Deirdre M. McCarthy
- To whom all correspondence should be
addressed: Deirdre M. McCarthy, Department of Biomedical Sciences, Florida State
University College of Medicine, 1115 West Call St., Tallahassee, FL 32306; Tele:
850-645-0263; Fax: 850-644-5781;
| | | | | |
Collapse
|
39
|
Jansson L, Louhivuori L, Wigren HK, Nordström T, Louhivuori V, Castrén M, Åkerman K. Brain-derived neurotrophic factor increases the motility of a particular N-methyl-d-aspartate /GABA-responsive subset of neural progenitor cells. Neuroscience 2012; 224:223-34. [DOI: 10.1016/j.neuroscience.2012.08.038] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 08/15/2012] [Accepted: 08/16/2012] [Indexed: 10/28/2022]
|
40
|
Yan BC, Park JH, Kim SK, Choi JH, Lee CH, Yoo KY, Kwon YG, Kim YM, Kim JD, Won MH. Comparison of trophic factors changes in the hippocampal CA1 region between the young and adult gerbil induced by transient cerebral ischemia. Cell Mol Neurobiol 2012; 32:1231-42. [PMID: 22552890 DOI: 10.1007/s10571-012-9848-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 04/16/2012] [Indexed: 10/28/2022]
Abstract
In the present study, we investigated neuronal death/damage in the gerbil hippocampal CA1 region (CA1) and compared changes in some trophic factors, such as brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF) and vascular endothelial growth factor (VEGF), in the CA1 between the adult and young gerbils after 5 min of transient cerebral ischemia. Most of pyramidal neurons (89%) were damaged 4 days after ischemia-reperfusion (I-R) in the adult; however, in the young, about 59% of pyramidal neurons were damaged 7 days after I-R. The immunoreactivity and levels of BDNF and VEGF, not GDNF, in the CA1 of the normal young were lower than those in the normal adult. Four days after I-R in the adult group, the immunoreactivity and levels of BDNF and VEGF were distinctively decreased, and the immunoreactivity and level of GDNF were increased. However, in the young group, all of their immunoreactivities and levels were much higher than those in the normal young group. From 7 days after I-R, all the immunoreactivities and levels were apparently decreased compared to those of the normal adult and young. In brief, we confirmed our recent finding: more delayed and less neuronal death occurred in the young following I-R, and we newly found that the immunoreactivities of trophic factors, such as BDNF, GDNF, and VEGF, in the stratum pyramidale of the CA1 in the young gerbil were much higher than those in the adult gerbil 4 days after transient cerebral ischemia.
Collapse
Affiliation(s)
- Bing Chun Yan
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 200-701, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
McCarthy DM, Gioioso V, Zhang X, Sharma N, Bhide PG. Neurogenesis and neuronal migration in the forebrain of the TorsinA knockout mouse embryo. Dev Neurosci 2012; 34:366-78. [PMID: 23018676 DOI: 10.1159/000342260] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 07/26/2012] [Indexed: 01/07/2023] Open
Abstract
Early-onset generalized torsion dystonia, also known as DYT1 dystonia, is a childhood onset heritable neurological movement disorder involving painful, involuntary muscle contractions, sustained abnormal postures, and repetitive movements. It is caused by a GAG deletion in the Tor1A gene located on chromosome 9. TorsinA, the product of the Tor1A gene, is expressed throughout the brain beginning early in embryonic development. It plays a role in the regulation of nuclear envelope-cytoskeletal interactions, and presumably nuclear translocation. Since nuclear translocation, powered by cytoskeletal traction, is critical for cell proliferation and migration, we examined whether neurogenesis and neuronal migration are affected in Tor1A-/- mouse brain. Our data show that interkinetic nuclear migration and the pattern of migration of newly generated neurons are impaired in the dorsal forebrain of the Tor1A-/- embryo. However, neurogenesis is not altered significantly. The rate of migration of cells from explants of the medial ganglionic eminence is also impaired in the Tor1A-/- embryo. Thus, loss of torsinA results in subtle but significant alterations in cell proliferation and migration in the embryonic forebrain. These subtle developmental changes are consistent with a lack of significant changes in neuronal numbers, neuronal positioning or size of brain regions in DYT1 dystonia patients.
Collapse
Affiliation(s)
- Deirdre M McCarthy
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32303, USA
| | | | | | | | | |
Collapse
|
42
|
Prenatal nicotine exposure mouse model showing hyperactivity, reduced cingulate cortex volume, reduced dopamine turnover, and responsiveness to oral methylphenidate treatment. J Neurosci 2012; 32:9410-8. [PMID: 22764249 DOI: 10.1523/jneurosci.1041-12.2012] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cigarette smoking, nicotine replacement therapy, and smokeless tobacco use during pregnancy are associated with cognitive disabilities later in life in children exposed prenatally to nicotine. The disabilities include attention deficit hyperactivity disorder (ADHD) and conduct disorder. However, the structural and neurochemical bases of these cognitive deficits remain unclear. Using a mouse model we show that prenatal nicotine exposure produces hyperactivity, selective decreases in cingulate cortical volume, and radial thickness, as well as decreased dopamine turnover in the frontal cortex. The hyperactivity occurs in both male and female offspring and peaks during the "active" or dark phase of the light/dark cycle. These features of the mouse model closely parallel the human ADHD phenotype, whether or not the ADHD is associated with prenatal nicotine exposure. A single oral, but not intraperitoneal, administration of a therapeutic equivalent dose (0.75 mg/kg) of methylphenidate decreases the hyperactivity and increases the dopamine turnover in the frontal cortex of the prenatally nicotine exposed mice, once again paralleling the therapeutic effects of this compound in ADHD subjects. Collectively, our data suggest that the prenatal nicotine exposure mouse model has striking parallels to the ADHD phenotype not only in behavioral, neuroanatomical, and neurochemical features, but also with respect to responsiveness of the behavioral phenotype to methylphenidate treatment. The behavioral, neurochemical, and anatomical biomarkers in the mouse model could be valuable for evaluating new therapies for ADHD and mechanistic investigations into its etiology.
Collapse
|
43
|
Kabir ZD, Lourenco F, Byrne ME, Katzman A, Lee F, Rajadhyaksha AM, Kosofsky BE. Brain-derived neurotrophic factor genotype impacts the prenatal cocaine-induced mouse phenotype. Dev Neurosci 2012; 34:184-97. [PMID: 22572518 DOI: 10.1159/000337712] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Accepted: 03/05/2012] [Indexed: 11/19/2022] Open
Abstract
Prenatal cocaine exposure leads to persistent alterations in the growth factor brain-derived neurotrophic factor (BDNF), particularly in the medial prefrontal cortex (mPFC) and hippocampus, brain regions important in cognitive functioning. BDNF plays an important role in the strengthening of existing synaptic connections as well as in the formation of new contacts during learning. A single nucleotide polymorphism in the BDNF gene (Val66Met), leading to a Met substitution for Val at codon 66 in the prodomain, is common in human populations, with an allele frequency of 20-30% in Caucasians. To study the interaction between prenatal cocaine exposure and BDNF, we have utilized a line of BDNF Val66Met transgenic mice on a Swiss Webster background in which BDNF(Met) is endogenously expressed. Examination of baseline levels of mature BDNF protein in the mPFC of prenatally cocaine-treated wild-type (Val66Val) and Val66Met mice revealed significantly lower levels compared to prenatally saline-treated mice. In contrast, in the hippocampus of prenatally saline- and cocaine-treated adult Val66Met mice, there were significantly lower levels of mature BDNF protein compared to Val66Val mice. In extinction of a conditioned fear, we found that prenatally cocaine-treated Val66Met mice had a deficit in recall of extinction. Examination of mature BDNF protein levels immediately after the test for extinction recall revealed lower levels in the mPFC of prenatally cocaine-treated Val66Met mice compared to saline-treated mice. However, 2 h after the extinction test, there was increased BDNF exons I, IV, and IX mRNA expression in the prelimbic cortex of the mPFC in the prenatally cocaine-treated BDNF Val66Met mice compared to prenatally saline-treated mice. Taken together, our results suggest the possibility that prenatal cocaine-induced constitutive alterations in BDNF mRNA and protein expression in the mPFC differentially poises animals for alterations in behaviorally induced gene activation, which are interactive with BDNF genotype and differentially impact those behaviors. Such findings in our prenatal cocaine mouse model suggest a gene X environment interaction of potential clinical relevance.
Collapse
Affiliation(s)
- Zeeba D Kabir
- Division of Pediatric Neurology, Department of Pediatrics, Weill Cornell Medical College, New York, NY 10021, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Stolp H, Neuhaus A, Sundramoorthi R, Molnár Z. The Long and the Short of it: Gene and Environment Interactions During Early Cortical Development and Consequences for Long-Term Neurological Disease. Front Psychiatry 2012; 3:50. [PMID: 22701439 PMCID: PMC3372875 DOI: 10.3389/fpsyt.2012.00050] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 05/01/2012] [Indexed: 01/21/2023] Open
Abstract
Cortical development is a complex amalgamation of proliferation, migration, differentiation, and circuit formation. These processes follow defined timescales and are controlled by a combination of intrinsic and extrinsic factors. It is currently unclear how robust and flexible these processes are and whether the developing brain has the capacity to recover from disruptions. What is clear is that there are a number of cognitive disorders or conditions that are elicited as a result of disrupted cortical development, although it may take a long time for the full pathophysiology of the conditions to be realized clinically. The critical window for the manifestation of a neurodevelopmental disorder is prolonged, and there is the potential for a complex interplay between genes and environment. While there have been extended investigations into the genetic basis of a number of neurological and mental disorders, limited definitive associations have been discovered. Many environmental factors, including inflammation and stress, have been linked to neurodevelopmental disorders, and it may be that a better understanding of the interplay between genes and environment will speed progress in this field. In particular, the development of the brain needs to be considered in the context of the whole materno-fetal unit as the degree of the metabolic, endocrine, or inflammatory responses, for example, will greatly influence the environment in which the brain develops. This review will emphasize the importance of extending neurodevelopmental studies to the contribution of the placenta, vasculature, cerebrospinal fluid, and to maternal and fetal immune response. These combined investigations are more likely to reveal genetic and environmental factors that influence the different stages of neuronal development and potentially lead to the better understanding of the etiology of neurological and mental disorders such as autism, epilepsy, cerebral palsy, and schizophrenia.
Collapse
Affiliation(s)
- Helen Stolp
- Department of Physiology, Anatomy and Genetics, University of Oxford Oxford, UK
| | | | | | | |
Collapse
|
45
|
McCarthy DM, Bhide PG. Prenatal Cocaine Exposure Decreases Parvalbumin-Immunoreactive Neurons and GABA-to-Projection Neuron Ratio in the Medial Prefrontal Cortex. Dev Neurosci 2012; 34:174-83. [DOI: 10.1159/000337172] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 02/07/2012] [Indexed: 11/19/2022] Open
|
46
|
Tropea TF, Kabir ZD, Kaur G, Rajadhyaksha AM, Kosofsky BE. Enhanced dopamine D1 and BDNF signaling in the adult dorsal striatum but not nucleus accumbens of prenatal cocaine treated mice. Front Psychiatry 2011; 2:67. [PMID: 22162970 PMCID: PMC3232639 DOI: 10.3389/fpsyt.2011.00067] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 11/12/2011] [Indexed: 01/18/2023] Open
Abstract
Previous work from our group and others utilizing animal models have demonstrated long-lasting structural and functional alterations in the meso-cortico-striatal dopamine pathway following prenatal cocaine (PCOC) treatment. We have shown that PCOC treatment results in augmented D1-induced cyclic AMP (cAMP) and cocaine-induced immediate-early gene expression in the striatum of adult mice. In this study we further examined basal as well as cocaine or D1-induced activation of a set of molecules known to be mediators of neuronal plasticity following psychostimulant treatment, with emphasis in the dorsal striatum (Str) and nucleus accumbens (NAc) of adult mice exposed to cocaine in utero. Basally, in the Str of PCOC treated mice there were significantly higher levels of (1) CREB and Ser133 P-CREB (2) Thr34 P-DARPP-32 and (3) GluA1 and Ser 845 P-GluA1 when compared to prenatal saline (PSAL) treated mice. In the NAc there were significantly higher basal levels of (1) CREB and Ser133 P-CREB, (2) Thr202/Tyr204 P-ERK2, and (3) Ser845 P-GluA1. Following acute administration of cocaine (15 mg/kg, i.p.) or D1 agonist (SKF 82958; 1 mg/kg, i.p.) there were significantly higher levels of Ser133 P-CREB, Thr34 P-DARPP-32, and Thr202/Tyr204 P-ERK2 in the Str that were evident in all animals tested. However, these cocaine-induced increases in phosphorylation were significantly augmented in PCOC mice compared to PSAL mice. In sharp contrast to the observations in the Str, in the NAc, acute administration of cocaine or D1 agonist significantly increased P-CREB and P-ERK2 in PSAL mice, a response that was not evident in PCOC mice. Examination of Ser 845 P-GluA1 revealed that cocaine or D1 agonist significantly increased levels in PSAL mice, but significantly decreased levels in the PCOC mice in both the Str and NAc. We also examined changes in brain-derived neurotrophic factor (BDNF). Our studies revealed significantly higher levels of the BDNF precursor, pro-BDNF, and one of its receptors, TrkB in the Str of PCOC mice compared to PSAL mice. These results suggest a persistent up-regulation of molecules critical to D1 and BDNF signaling in the Str of adult mice exposed to cocaine in utero. These molecular adaptations may underlie components of the behavioral deficits evident in exposed animals and a subset of exposed humans, and may represent a therapeutic target for ameliorating aspects of the PCOC-induced phenotype.
Collapse
Affiliation(s)
- Thomas F. Tropea
- Division of Pediatric Neurology, Department of Pediatrics, Weill Cornell Medical CollegeNew York, NY, USA
- College of Osteopathic Medicine, University of New EnglandBiddeford, ME, USA
| | - Zeeba D. Kabir
- Division of Pediatric Neurology, Department of Pediatrics, Weill Cornell Medical CollegeNew York, NY, USA
- Graduate Program in Neurosciences, Weill Cornell Medical CollegeNew York, NY, USA
| | - Gagandeep Kaur
- School of Environmental and Biological Sciences, Rutgers, The State University of New JerseyNew Brunswick, NJ, USA
| | - Anjali M. Rajadhyaksha
- Division of Pediatric Neurology, Department of Pediatrics, Weill Cornell Medical CollegeNew York, NY, USA
- Graduate Program in Neurosciences, Weill Cornell Medical CollegeNew York, NY, USA
| | - Barry E. Kosofsky
- Division of Pediatric Neurology, Department of Pediatrics, Weill Cornell Medical CollegeNew York, NY, USA
- Graduate Program in Neurosciences, Weill Cornell Medical CollegeNew York, NY, USA
| |
Collapse
|