1
|
Mihalj D, Bukatova S, Reichova A, Havranek T, Bacova Z, Szeiffova Bacova B, Bakos J. Developmental effects of oxytocin on GABAergic neurons in the olfactory brain regions. Neuroscience 2024; 555:184-193. [PMID: 39094821 DOI: 10.1016/j.neuroscience.2024.07.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/17/2024] [Accepted: 07/21/2024] [Indexed: 08/04/2024]
Abstract
Oxytocin affects social recognition, interactions, and behavior in adults. Despite growing data on the role of oxytocin in the sensory systems, its effects on early olfactory system development remain poorly understood. The present study aimed to investigate the developmental impact of oxytocin on selected parameters of the GABAergic system in olfactory brain regions. We found a significant increase in the expression of GABAergic markers and scaffolding proteins in the olfactory bulb during the early stages of development in both male and female rats, regardless of oxytocin treatment administered on postnatal days 2 and 3 (P2 and P3, 5 µg/pup). Oxytocin administration markedly reduced the expression of the scaffolding protein Gephyrin in male rats and it led to a significant increase in the number of GABAergic synaptic puncta in the piriform cortex of male rats at P5, P7, and P9. Our data suggest that the developmental action of oxytocin in relation to the GABAergic system may represent a mechanism by which the plasticity and maturation of olfactory brain regions are regulated.
Collapse
Affiliation(s)
- Denisa Mihalj
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Stanislava Bukatova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Alexandra Reichova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Tomas Havranek
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia; Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Zuzana Bacova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Barbara Szeiffova Bacova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Jan Bakos
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia; Faculty of Medicine, Comenius University, Bratislava, Slovakia.
| |
Collapse
|
2
|
Liwang JK, Kronman FA, Minteer JA, Wu YT, Vanselow DJ, Ben-Simon Y, Taormina M, Parmaksiz D, Way SW, Zeng H, Tasic B, Ng L, Kim Y. epDevAtlas: Mapping GABAergic cells and microglia in postnatal mouse brains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.24.568585. [PMID: 38045330 PMCID: PMC10690281 DOI: 10.1101/2023.11.24.568585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
During development, brain regions follow encoded growth trajectories. Compared to classical brain growth charts, high-definition growth charts could quantify regional volumetric growth and constituent cell types, improving our understanding of typical and pathological brain development. Here, we create high-resolution 3D atlases of the early postnatal mouse brain, using Allen CCFv3 anatomical labels, at postnatal days (P) 4, 6, 8, 10, 12, and 14, and determine the volumetric growth of different brain regions. We utilize 11 different cell type-specific transgenic animals to validate and refine anatomical labels. Moreover, we reveal region-specific density changes in γ-aminobutyric acid-producing (GABAergic), cortical layer-specific cell types, and microglia as key players in shaping early postnatal brain development. We find contrasting changes in GABAergic neuronal densities between cortical and striatal areas, stabilizing at P12. Moreover, somatostatin-expressing cortical interneurons undergo regionally distinct density reductions, while vasoactive intestinal peptide-expressing interneurons show no significant changes. Remarkably, microglia transition from high density in white matter tracks to gray matter at P10, and show selective density increases in sensory processing areas that correlate with the emergence of individual sensory modalities. Lastly, we create an open-access web-visualization ( https://kimlab.io/brain-map/epDevAtlas ) for cell-type growth charts and developmental atlases for all postnatal time points.
Collapse
|
3
|
Massri AJ, Fitzpatrick M, Cunny H, Li JL, Harry GJ. Differential gene expression profiling implicates altered network development in rat postnatal day 4 cortex following 4-Methylimidazole (4-MeI) induced maternal seizures. Neurotoxicol Teratol 2023; 100:107301. [PMID: 37783441 PMCID: PMC10843020 DOI: 10.1016/j.ntt.2023.107301] [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: 06/23/2023] [Revised: 08/31/2023] [Accepted: 09/19/2023] [Indexed: 10/04/2023]
Abstract
Compromised maternal health leading to maternal seizures can have adverse effects on the healthy development of offspring. This may be the result of inflammation, hypoxia-ischemia, and altered GABA signaling. The current study examined cortical tissue from F2b (2nd litter of the 2nd generation) postnatal day 4 (PND4) offspring of female Harlan SD rats chronically exposed to the seizuregenic compound, 4-Methylimidazole (0, 750, or 2500 ppm 4-MeI). Maternal seizures were evident only at 2500 ppm 4-MeI. GABA related gene expression as examined by qRT-PCR and whole genome microarray showed no indication of disrupted GABA or glutamatergic signaling. Canonical pathway hierarchical clustering and multi-omics combinatory genomic (CNet) plots of differentially expressed genes (DEG) showed alterations in genes associated with regulatory processes of cell development including neuronal differentiation and synaptogenesis. Functional enrichment analysis showed a similarity of cellular processes across the two exposure groups however, the genes comprising each cluster were primarily unique rather than shared and often showed different directionality. A dose-related induction of cytokine signaling was indicated however, pathways associated with individual cytokine signaling were not elevated, suggesting an alternative involvement of cytokine signaling. Pathways related to growth process and cell signaling showed a negative activation supporting an interpretation of disruption or delay in developmental processes at the 2500 ppm 4-MeI exposure level with maternal seizures. Thus, while GABA signaling was not altered as has been observed with maternal seizures, the pattern of DEG suggested a potential for alteration in neuronal network formation.
Collapse
Affiliation(s)
- Abdull J Massri
- Integrative Bioinformatics, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Mackenzie Fitzpatrick
- Mechanistic Toxicology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Helen Cunny
- Office of the Scientific Director, Division of Translational Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Jian-Liang Li
- Integrative Bioinformatics, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - G Jean Harry
- Mechanistic Toxicology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
4
|
Developmental Inhibitory Changes in the Primary Somatosensory Cortex of the Stargazer Mouse Model of Absence Epilepsy. Biomolecules 2023; 13:biom13010186. [PMID: 36671571 PMCID: PMC9856073 DOI: 10.3390/biom13010186] [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: 12/18/2022] [Revised: 01/08/2023] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
Childhood absence epilepsy seizures arise in the cortico-thalamocortical network due to multiple cellular and molecular mechanisms, which are still under investigation. Understanding the precise mechanisms is imperative given that treatment fails in ~30% of patients while adverse neurological sequelae remain common. Impaired GABAergic neurotransmission is commonly reported in research models investigating these mechanisms. Recently, we reported a region-specific reduction in the whole-tissue and synaptic GABAA receptor (GABAAR) α1 subunit and an increase in whole-tissue GAD65 in the primary somatosensory cortex (SoCx) of the adult epileptic stargazer mouse compared with its non-epileptic (NE) littermate. The current study investigated whether these changes occurred prior to the onset of seizures on postnatal days (PN) 17-18, suggesting a causative role. Synaptic and cytosolic fractions were biochemically isolated from primary SoCx lysates followed by semiquantitative Western blot analyses for GABAAR α1 and GAD65. We found no significant changes in synaptic GABAAR α1 and cytosolic GAD65 in the primary SoCx of the stargazer mice at the critical developmental stages of PN 7-9, 13-15, and 17-18. This indicates that altered levels of GABAAR α1 and GAD65 in adult mice do not directly contribute to the initial onset of absence seizures but are a later consequence of seizure activity.
Collapse
|
5
|
Crombie GK, Palliser HK, Shaw JC, Hodgson DM, Walker DW, Hirst JJ. Effects of prenatal stress on behavioural and neurodevelopmental outcomes are altered by maternal separation in the neonatal period. Psychoneuroendocrinology 2021; 124:105060. [PMID: 33333379 DOI: 10.1016/j.psyneuen.2020.105060] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/08/2020] [Accepted: 11/09/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND Chronic psychosocial stress during pregnancy and/or after birth, and the associated elevation in cortisol, is linked with the onset of behavioural disorders in childhood. Previously, prenatal stress has been shown to reduce neurosteroid pathways in the fetus and the levels of the neurosteroid and GABAA receptor agonist, allopregnanolone. In late gestation, elevated levels of GABAergic activity increases inhibitory tone and protects against excessive excitation. These levels of allopregnanolone may also contribute to promoting myelination, thus stress-induced suppression of protective neurosteroid levels may disrupt neurodevelopmental processes and can result in reduced myelination. The objective of this study was to examine whether prenatal and postnatal stress reduces levels of inhibitory pathways to result in behavioural, myelin, and GABAergic/glutamatergic pathway deficits in the hippocampus at a postnatal time point in the guinea pig equivalent to childhood in humans. METHODS Pregnant guinea pig dams were exposed to prenatal stress (PRE) with strobe light exposure for 2 h/day on gestational age (GA) 50, 55, 60 and 65 (term is ∼GA70), with postnatal stress (POST) caused by maternal separation for 2 h/day from postnatal day (PND) 1-7), or a double-hit of both stressors (PRE + POST). Control dams and offspring groups (CON) were handled at the same time each day without causing stress. Behavioural outcomes were assessed using open field and elevated plus maze testing on PND27. After euthanasia on PND30, plasma samples were collected for steroid quantification of cortisol, allopregnanolone and progesterone by ELISA. Hippocampal samples were collected to assess markers of oligodendrocyte development and mature cells by myelin basic protein (MBP) immunostaining and GABAergic and glutamatergic pathway component gene expression by real time PCR. RESULTS Male guinea pig offspring exposed to prenatal stress exhibited hyperactive-like behaviour at childhood equivalence, while female offspring displayed anxious-like behaviour, to a lesser extent. In both sexes, MBP immunostaining was significantly decreased in the hippocampal region following prenatal stress, despite normal levels of MBP mRNA, which suggests a disruption to the MBP protein translation pathway. Many components of the GABAergic and glutamatergic pathways were disrupted following prenatal stress, notably GABAA receptor subunits, GABA production and uptake, glutamate ionotropic and metabotropic receptor subunits and glutamate transport. Following prenatal + postnatal stress, many of the behavioural and neurodevelopmental deficits were improved compared to the prenatal stress only group. CONCLUSION We conclude that prenatal stress disrupts GABAergic and glutamatergic pathways that may contribute to reduced myelination and subsequent behavioural deficits in the offspring. The deficits seen following prenatal stress are ameliorated when paired with subsequent postnatal stress, which highlights the early postnatal period as an important treatment window.
Collapse
Affiliation(s)
- Gabrielle K Crombie
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, NSW, Australia; School of Biomedical Sciences and Pharmacy, University of Newcastle, NSW, Australia.
| | - Hannah K Palliser
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, NSW, Australia; School of Biomedical Sciences and Pharmacy, University of Newcastle, NSW, Australia
| | - Julia C Shaw
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, NSW, Australia; School of Biomedical Sciences and Pharmacy, University of Newcastle, NSW, Australia
| | | | - David W Walker
- School of Health and Biomedical Sciences, RMIT University, VIC, Australia
| | - Jonathan J Hirst
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, NSW, Australia; School of Biomedical Sciences and Pharmacy, University of Newcastle, NSW, Australia
| |
Collapse
|
6
|
Willis A, Pratt JA, Morris BJ. BDNF and JNK Signaling Modulate Cortical Interneuron and Perineuronal Net Development: Implications for Schizophrenia-Linked 16p11.2 Duplication Syndrome. Schizophr Bull 2020; 47:812-826. [PMID: 33067994 PMCID: PMC8084442 DOI: 10.1093/schbul/sbaa139] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Schizophrenia (SZ) is a neurodevelopmental disorder caused by the interaction of genetic and environmental risk factors. One of the strongest genetic risk variants is duplication (DUP) of chr.16p11.2. SZ is characterized by cortical gamma-amino-butyric acid (GABA)ergic interneuron dysfunction and disruption to surrounding extracellular matrix structures, perineuronal nets (PNNs). Developmental maturation of GABAergic interneurons, and also the resulting closure of the critical period of cortical plasticity, is regulated by brain-derived neurotrophic factor (BDNF), although the mechanisms involved are unknown. Here, we show that BDNF promotes GABAergic interneuron and PNN maturation through JNK signaling. In mice reproducing the 16p11.2 DUP, where the JNK upstream activator Taok2 is overexpressed, we find that JNK is overactive and there are developmental abnormalities in PNNs, which persist into adulthood. Prefrontal cortex parvalbumin (PVB) expression is reduced, while PNN intensity is increased. Additionally, we report a unique role for TAOK2 signaling in the regulation of PVB interneurons. Our work implicates TAOK2-JNK signaling in cortical interneuron and PNN development, and in the responses to BDNF. It also demonstrates that over-activation of this pathway in conditions associated with SZ risk causes long-lasting disruption in cortical interneurons.
Collapse
Affiliation(s)
- Ashleigh Willis
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland, UK
| | - Judith A Pratt
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Brian J Morris
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland, UK,To whom correspondence should be addressed; Institute of Neuroscience and Psychology, University of Glasgow, G12 8QQ, Glasgow, Scotland, UK; tel: 0044-141-330-5361, fax: 0044-141-330-5659, e-mail:
| |
Collapse
|
7
|
Dade M, Berzero G, Izquierdo C, Giry M, Benazra M, Delattre JY, Psimaras D, Alentorn A. Neurological Syndromes Associated with Anti-GAD Antibodies. Int J Mol Sci 2020; 21:E3701. [PMID: 32456344 PMCID: PMC7279468 DOI: 10.3390/ijms21103701] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 12/11/2022] Open
Abstract
Glutamic acid decarboxylase (GAD) is an intracellular enzyme whose physiologic function is the decarboxylation of glutamate to gamma-aminobutyric acid (GABA), the main inhibitory neurotransmitter within the central nervous system. GAD antibodies (Ab) have been associated with multiple neurological syndromes, including stiff-person syndrome, cerebellar ataxia, and limbic encephalitis, which are all considered to result from reduced GABAergic transmission. The pathogenic role of GAD Ab is still debated, and some evidence suggests that GAD autoimmunity might primarily be cell-mediated. Diagnosis relies on the detection of high titers of GAD Ab in serum and/or in the detection of GAD Ab in the cerebrospinal fluid. Due to the relative rarity of these syndromes, treatment schemes and predictors of response are poorly defined, highlighting the unmet need for multicentric prospective trials in this population. Here, we reviewed the main clinical characteristics of neurological syndromes associated with GAD Ab, focusing on pathophysiologic mechanisms.
Collapse
Affiliation(s)
- Maëlle Dade
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service de Neurologie 2-Mazarin, 75013 Paris, France; (M.D.); (G.B.); (J.-Y.D.); (D.P.)
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, 75013 Paris, France; (M.G.); (M.B.)
| | - Giulia Berzero
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service de Neurologie 2-Mazarin, 75013 Paris, France; (M.D.); (G.B.); (J.-Y.D.); (D.P.)
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, 75013 Paris, France; (M.G.); (M.B.)
- Neuroncology Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Cristina Izquierdo
- Department of Neuroscience, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain;
| | - Marine Giry
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, 75013 Paris, France; (M.G.); (M.B.)
| | - Marion Benazra
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, 75013 Paris, France; (M.G.); (M.B.)
| | - Jean-Yves Delattre
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service de Neurologie 2-Mazarin, 75013 Paris, France; (M.D.); (G.B.); (J.-Y.D.); (D.P.)
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, 75013 Paris, France; (M.G.); (M.B.)
| | - Dimitri Psimaras
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service de Neurologie 2-Mazarin, 75013 Paris, France; (M.D.); (G.B.); (J.-Y.D.); (D.P.)
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, 75013 Paris, France; (M.G.); (M.B.)
| | - Agusti Alentorn
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Service de Neurologie 2-Mazarin, 75013 Paris, France; (M.D.); (G.B.); (J.-Y.D.); (D.P.)
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, 75013 Paris, France; (M.G.); (M.B.)
| |
Collapse
|
8
|
Navabpour S, Kwapis JL, Jarome TJ. A neuroscientist's guide to transgenic mice and other genetic tools. Neurosci Biobehav Rev 2020; 108:732-748. [PMID: 31843544 PMCID: PMC8049509 DOI: 10.1016/j.neubiorev.2019.12.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/05/2019] [Accepted: 12/09/2019] [Indexed: 12/14/2022]
Abstract
The past decade has produced an explosion in the number and variety of genetic tools available to neuroscientists, resulting in an unprecedented ability to precisely manipulate the genome and epigenome in behaving animals. However, no single resource exists that describes all of the tools available to neuroscientists. Here, we review the genetic, transgenic, and viral techniques that are currently available to probe the complex relationship between genes and cognition. Topics covered include types of traditional transgenic mouse models (knockout, knock-in, reporter lines), inducible systems (Cre-loxP, Tet-On, Tet-Off) and cell- and circuit-specific systems (TetTag, TRAP, DIO-DREADD). Additionally, we provide details on virus-mediated and siRNA/shRNA approaches, as well as a comprehensive discussion of the myriad manipulations that can be made using the CRISPR-Cas9 system, including single base pair editing and spatially- and temporally-regulated gene-specific transcriptional control. Collectively, this review will serve as a guide to assist neuroscientists in identifying and choosing the appropriate genetic tools available to study the complex relationship between the brain and behavior.
Collapse
Affiliation(s)
- Shaghayegh Navabpour
- Fralin Biomedical Research Institute, Translational Biology, Medicine and Health, Roanoke, VA, USA
| | - Janine L Kwapis
- Department of Biology, Pennsylvania State University, College Park, PA, USA; Center for the Molecular Investigation of Neurological Disorders (CMIND), Pennsylvania State University, College Park, PA, USA.
| | - Timothy J Jarome
- Fralin Biomedical Research Institute, Translational Biology, Medicine and Health, Roanoke, VA, USA; Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA; School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
| |
Collapse
|
9
|
Thorpe HHA, Hamidullah S, Jenkins BW, Khokhar JY. Adolescent neurodevelopment and substance use: Receptor expression and behavioral consequences. Pharmacol Ther 2019; 206:107431. [PMID: 31706976 DOI: 10.1016/j.pharmthera.2019.107431] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2019] [Indexed: 12/18/2022]
Abstract
Adolescence is the transitional period between childhood and adulthood, during which extensive brain development occurs. Since this period also overlaps with the initiation of drug use, it is important to consider how substance use during this time might produce long-term neurobiological alterations, especially against the backdrop of developmental changes in neurotransmission. Alcohol, cannabis, nicotine, and opioids all produce marked changes in the expression and function of the neurotransmitter and receptor systems with which they interact. These acute and chronic alterations also contribute to behavioral consequences ranging from increased addiction risk to cognitive or neuropsychiatric behavioral dysfunctions. The current review provides an in-depth overview and update of the developmental changes in neurotransmission during adolescence, as well as the impact of drug exposure during this neurodevelopmental window. While most of these factors have been studied in animal models, which are the focus of this review, future longitudinal studies in humans that assess neural function and behavior will help to confirm pre-clinical findings. Furthermore, the neural changes induced by each drug should also be considered in the context of other contributing factors, such as sex. Further understanding of these consequences can help in the identification of novel approaches for preventing and reversing the neurobiological effects of adolescent substance use.
Collapse
Affiliation(s)
- Hayley H A Thorpe
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Ontario, Canada
| | - Shahnaza Hamidullah
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Ontario, Canada
| | - Bryan W Jenkins
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Ontario, Canada
| | - Jibran Y Khokhar
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Ontario, Canada.
| |
Collapse
|
10
|
Abstract
Abstract
Introduction: GAD2 gene encodes the glutamate decarboxylase enzyme which catalyses the transformation of glutamate into γ-aminobutyric acid, GABA. It is suggested that some polymorphic alleles of GAD2 gene, such as -243A>G, have an increased transcriptional effect compared with the wild type, which results in an increase of GABA in the hypothalamus with the subsequent increase of the neuropeptide Y, thus exacerbating the hunger centre and the appetite. The aim of this study was to observe an association between the -243A>G polymorphism with obesity, comparatively studying a group of obese patients and a group of patients with normal weight.
Patients and method: 127 patients were clinically evaluated in the Genetic and Endocrine Department of Children’s Emergency Clinical Hospital, Cluj. The patients were included in two study groups, case group, with obesity (BMI higher than 97 kg/m2) and control group, with normal weight (BMI less than 97 kg/m2). Genotyping for GAD2-243A>G polymorphism was performed using PCR-RFLP technique, the two groups being compared regarding the genotypes and phenotypes.
Results and conclusions: In the obesity group, there is a statistically significant difference in BMI (kg/m2) between the subgroups with different genotypes (p=0.01), the AA genotype being less severely affected than AG and GG genotypes. In the normal weight group there is no association between BMI and different genotypes (AA, AG or GG). Also, there is a greater distribution of GG genotypes and G allele in the obesity group compared with the control group, with an odds ratio which suggest that -243A>G polymorphism is a risk factor in obesity development (GG genotype OR=3.76, G allele OR=1.73, p=0.04).
The finding of our study is important in explaining the multifactorial model of obesity, our research demonstrating that the GAD2-243 A> G variant could be a risk factor that added to other obesogenic factors would potentiate their effect.
Collapse
|
11
|
Bessières B, Jia M, Travaglia A, Alberini CM. Developmental changes in plasticity, synaptic, glia, and connectivity protein levels in rat basolateral amygdala. ACTA ACUST UNITED AC 2019; 26:436-448. [PMID: 31615855 PMCID: PMC6796789 DOI: 10.1101/lm.049866.119] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 08/09/2019] [Indexed: 01/01/2023]
Abstract
The basolateral complex of amygdala (BLA) processes emotionally arousing aversive and rewarding experiences. The BLA is critical for acquisition and storage of threat-based memories and the modulation of the consolidation of arousing explicit memories, that is, the memories that are encoded and stored by the medial temporal lobe. In addition, in conjunction with the medial prefrontal cortex (mPFC), the BLA plays an important role in fear memory extinction. The BLA develops relatively early in life, but little is known about the molecular changes that accompany its development. Here, we quantified relative basal expression levels of sets of plasticity, synaptic, glia, and connectivity proteins in the rat BLA at various developmental ages: postnatal day 17 (PN17, infants), PN24 (juveniles), and PN80 (young adults). We found that the levels of activation markers of brain plasticity, including phosphorylation of CREB at Ser133, CamKIIα at Thr286, pERK1/pERK2 at Thr202/Tyr204, and GluA1 at Ser831 and Ser845, were significantly higher in infant and juvenile compared with adult brain. In contrast, age increase was accompanied by a significant augmentation in the levels of proteins that mark synaptogenesis and synapse maturation, such as synaptophysin, PSD95, SynCAM, GAD65, GAD67, and GluN2A/GluN2B ratio. Finally, we observed significant age-associated changes in structural markers, including MAP2, MBP, and MAG, suggesting that the structural connectivity of the BLA increases over time. The biological differences in the BLA between developmental ages compared with adulthood suggest the need for caution in extrapolating conclusions based on BLA-related brain plasticity and behavioral studies conducted at different developmental stages.
Collapse
Affiliation(s)
- Benjamin Bessières
- Center for Neural Science, New York University, New York, New York 10003, USA
| | - Margaret Jia
- Center for Neural Science, New York University, New York, New York 10003, USA
| | - Alessio Travaglia
- Center for Neural Science, New York University, New York, New York 10003, USA
| | - Cristina M Alberini
- Center for Neural Science, New York University, New York, New York 10003, USA
| |
Collapse
|
12
|
Gluncic V, Moric M, Chu Y, Hanko V, Li J, Lukić IK, Lukić A, Edassery SL, Kroin JS, Persons AL, Perry P, Kelly L, Shiveley TJ, Nice K, Napier CT, Kordower JH, Tuman KJ. In utero Exposure to Anesthetics Alters Neuronal Migration Pattern in Developing Cerebral Cortex and Causes Postnatal Behavioral Deficits in Rats. Cereb Cortex 2019; 29:5285-5301. [DOI: 10.1093/cercor/bhz065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Abstract
During fetal development, cerebral cortical neurons are generated in the proliferative zone along the ventricles and then migrate to their final positions. To examine the impact of in utero exposure to anesthetics on neuronal migration, we injected pregnant rats with bromodeoxyuridine to label fetal neurons generated at embryonic Day (E) 17 and then randomized these rats to 9 different groups receiving 3 different means of anesthesia (oxygen/control, propofol, isoflurane) for 3 exposure durations (20, 50, 120 min). Histological analysis of brains from 54 pups revealed that significant number of neurons in anesthetized animals failed to acquire their correct cortical position and remained dispersed within inappropriate cortical layers and/or adjacent white matter. Behavioral testing of 86 littermates pointed to abnormalities that correspond to the aberrations in the brain areas that are specifically developing during the E17. In the second set of experiments, fetal brains exposed to isoflurane at E16 had diminished expression of the reelin and glutamic acid decarboxylase 67, proteins critical for neuronal migration. Together, these results call for cautious use of anesthetics during the neuronal migration period in pregnancy and more comprehensive investigation of neurodevelopmental consequences for the fetus and possible consequences later in life.
Collapse
Affiliation(s)
- V Gluncic
- Department of Anesthesiology, Rush University Medical Center, Chicago, IL, USA
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago IL, USA
| | - M Moric
- Department of Anesthesiology, Rush University Medical Center, Chicago, IL, USA
| | - Y Chu
- Department of Neurological Sciences, Rush Medical College, Rush University Medical Center, Chicago, IL, USA
| | - V Hanko
- Department of Anesthesiology, Rush University Medical Center, Chicago, IL, USA
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - J Li
- Department of Anesthesiology, Rush University Medical Center, Chicago, IL, USA
| | - I K Lukić
- Department of Anesthesiology, Rush University Medical Center, Chicago, IL, USA
| | - A Lukić
- Department of Anesthesiology, Rush University Medical Center, Chicago, IL, USA
| | - S L Edassery
- Department of Pharmacology, Rush Medical College, Rush University Medical Center, Chicago, IL, USA
| | - J S Kroin
- Department of Anesthesiology, Rush University Medical Center, Chicago, IL, USA
| | - A L Persons
- Department of Pharmacology, Rush Medical College, Rush University Medical Center, Chicago, IL, USA
- The Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, USA
| | - P Perry
- Department of Anesthesiology, Rush University Medical Center, Chicago, IL, USA
| | - L Kelly
- Department of Neurological Sciences, Rush Medical College, Rush University Medical Center, Chicago, IL, USA
| | - T J Shiveley
- Department of Anesthesiology, Rush University Medical Center, Chicago, IL, USA
| | - K Nice
- Department of Neurological Sciences, Rush Medical College, Rush University Medical Center, Chicago, IL, USA
| | - C T Napier
- Department of Pharmacology, Rush Medical College, Rush University Medical Center, Chicago, IL, USA
- The Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, USA
- Department of Psychiatry, Rush Medical College, Rush University Medical Center, Chicago, IL, USA
| | - J H Kordower
- Department of Neurological Sciences, Rush Medical College, Rush University Medical Center, Chicago, IL, USA
| | - K J Tuman
- Department of Anesthesiology, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
13
|
Batista G, Hensch TK. Critical Period Regulation by Thyroid Hormones: Potential Mechanisms and Sex-Specific Aspects. Front Mol Neurosci 2019; 12:77. [PMID: 31024251 PMCID: PMC6461013 DOI: 10.3389/fnmol.2019.00077] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/11/2019] [Indexed: 12/24/2022] Open
Abstract
Adequate perinatal levels of thyroid hormones (THs) are required for normal brain function and development. Studies in non-mammalian species suggest that TH might be involved in the regulation of critical periods (CPs) of heightened plasticity. Yet, it is largely unknown what mechanisms controlling such CPs might be under TH regulation. Here, we briefly review the influence of TH in early life across evolution. We discuss possible links between TH and known circuit and/or molecular mechanisms determining the timing of CPs of heightened brain plasticity. We focus on the role of parvalbumin-positive (PV) interneurons since their maturation defines CP onset and closure. Specifically, abnormal PV circuits are associated with low perinatal levels of TH, possibly because thyroid hypofunction may increase oxidative stress and/or dysregulate Otx2-mediated maturation of neuroprotective perineuronal nets. In addition, the level of cholinergic transmission is important for CP plasticity. Potentially, TH levels could affect gain changes in cholinergic transmission that can alter brain development. We believe that understanding how TH impacts CPs of circuit refinement will shed light onto the principles underlying normal developmental trajectories. Given that the thyroid gland expresses estrogen and androgen receptors, its activity can potentially be regulated differently between the sexes, contributing to sexually dimorphic behaviors.
Collapse
Affiliation(s)
- Gervasio Batista
- Center for Brain Science, Department of Molecular Cellular Biology, Harvard University, Cambridge, MA, United States
| | - Takao K Hensch
- Center for Brain Science, Department of Molecular Cellular Biology, Harvard University, Cambridge, MA, United States.,FM Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Boston, MA, United States.,International Research Center for Neurointelligence, University of Tokyo Institutes for Advanced Study, Tokyo, Japan
| |
Collapse
|
14
|
Carmona-Alcocer V, Rohr KE, Joye DAM, Evans JA. Circuit development in the master clock network of mammals. Eur J Neurosci 2018; 51:82-108. [PMID: 30402923 DOI: 10.1111/ejn.14259] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/08/2018] [Accepted: 10/31/2018] [Indexed: 12/24/2022]
Abstract
Daily rhythms are generated by the circadian timekeeping system, which is orchestrated by the master circadian clock in the suprachiasmatic nucleus (SCN) of mammals. Circadian timekeeping is endogenous and does not require exposure to external cues during development. Nevertheless, the circadian system is not fully formed at birth in many mammalian species and it is important to understand how SCN development can affect the function of the circadian system in adulthood. The purpose of the current review is to discuss the ontogeny of cellular and circuit function in the SCN, with a focus on work performed in model rodent species (i.e., mouse, rat, and hamster). Particular emphasis is placed on the spatial and temporal patterns of SCN development that may contribute to the function of the master clock during adulthood. Additional work aimed at decoding the mechanisms that guide circadian development is expected to provide a solid foundation upon which to better understand the sources and factors contributing to aberrant maturation of clock function.
Collapse
Affiliation(s)
| | - Kayla E Rohr
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| | - Deborah A M Joye
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| | - Jennifer A Evans
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| |
Collapse
|
15
|
Magri C, Giacopuzzi E, La Via L, Bonini D, Ravasio V, Elhussiny MEA, Orizio F, Gangemi F, Valsecchi P, Bresciani R, Barbon A, Vita A, Gennarelli M. A novel homozygous mutation in GAD1 gene described in a schizophrenic patient impairs activity and dimerization of GAD67 enzyme. Sci Rep 2018; 8:15470. [PMID: 30341396 PMCID: PMC6195539 DOI: 10.1038/s41598-018-33924-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 10/08/2018] [Indexed: 12/30/2022] Open
Abstract
Recently, by whole exome sequencing of schizophrenia (SCZ) patients, we identified a subject that was homozygous for a novel missense substitution (c.391 A > G) in the glutamate acid decarboxylase 1 (GAD1) gene. GAD1 encodes for GAD67 enzyme, catalyzing the production of gamma-aminobutyric acid (GABA) from L-glutamic acid. Here, we studied the impact of this mutation on GAD67 activity, dimerization and subcellular localization. Biochemical assay revealed that c.391 A > G reduces GAD67 enzymatic activity by ~30%, probably due to the impaired homodimerization of homozygous mutants as highlighted by proximity ligation assays. The mutational screening of 120 genes of the "GABAergic system" in a cohort of 4,225 SCZ cases and 5,834 controls (dbGaP: phs000473.v1.p2), did not identify other cases that were homozygous for ultra-rare variants in GAD1, but highlighted an increased frequency of cases that were homozygous for rare variants in genes of the GABA system (SCZ: 0.14% vs. Controls: 0.00%; p-value = 0.0055). In conclusion, this study demonstrates the functional impact of c.391 A > G variant and its biological effect makes it a good candidate as risk variant for SCZ. This study also supports an involvement of ultra-rare variants in GABAergic genes in the etiopathogenesis of SCZ.
Collapse
Affiliation(s)
- Chiara Magri
- Unit of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Edoardo Giacopuzzi
- Unit of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luca La Via
- Unit of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Daniela Bonini
- Unit of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Viola Ravasio
- Unit of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Mohammed E A Elhussiny
- Unit of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Flavia Orizio
- Unit of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Fabrizio Gangemi
- Unit of Physics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Paolo Valsecchi
- Neuroscience Section, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Department of Mental Health, Spedali Civili Hospital, Brescia, Italy
| | - Roberto Bresciani
- Unit of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Alessandro Barbon
- Unit of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Antonio Vita
- Neuroscience Section, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Department of Mental Health, Spedali Civili Hospital, Brescia, Italy
| | - Massimo Gennarelli
- Unit of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Genetic Unit, IRCCS Centro S. Giovanni di Dio Fatebenefratelli, Brescia, Italy
| |
Collapse
|
16
|
Jia M, Travaglia A, Pollonini G, Fedele G, Alberini CM. Developmental changes in plasticity, synaptic, glia, and connectivity protein levels in rat medial prefrontal cortex. ACTA ACUST UNITED AC 2018; 25:533-543. [PMID: 30224556 PMCID: PMC6149953 DOI: 10.1101/lm.047753.118] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/06/2018] [Indexed: 12/22/2022]
Abstract
The medial prefrontal cortex (mPFC) plays a critical role in complex brain functions including decision-making, integration of emotional, and cognitive aspects in memory processing and memory consolidation. Because relatively little is known about the molecular mechanisms underlying its development, we quantified rat mPFC basal expression levels of sets of plasticity, synaptic, glia, and connectivity proteins at different developmental ages. Specifically, we compared the mPFC of rats at postnatal day 17 (PN17), when they are still unable to express long-term contextual and spatial memories, to rat mPFC at PN24, when they have acquired the ability of long-term memory expression and finally to the mPFC of adult rats. We found that, with increased age, there are remarkable and significant decreases in markers of cell activation and significant increases in proteins that mark synaptogenesis and synapse maturation. Furthermore, we found significant changes in structural markers over the ages, suggesting that structural connectivity of the mPFC increases over time. Finally, the substantial biological difference in mPFC at different ages suggest caution in extrapolating conclusions from brain plasticity studies conducted at different developmental stages.
Collapse
Affiliation(s)
- Margaret Jia
- Center for Neural Science, New York University, New York, New York 10003, USA
| | - Alessio Travaglia
- Center for Neural Science, New York University, New York, New York 10003, USA
| | - Gabriella Pollonini
- Center for Neural Science, New York University, New York, New York 10003, USA
| | - Giuseppe Fedele
- Center for Neural Science, New York University, New York, New York 10003, USA
| | - Cristina M Alberini
- Center for Neural Science, New York University, New York, New York 10003, USA
| |
Collapse
|
17
|
Tao R, Davis KN, Li C, Shin JH, Gao Y, Jaffe AE, Gondré-Lewis MC, Weinberger DR, Kleinman JE, Hyde TM. GAD1 alternative transcripts and DNA methylation in human prefrontal cortex and hippocampus in brain development, schizophrenia. Mol Psychiatry 2018; 23:1496-1505. [PMID: 28485403 PMCID: PMC7564279 DOI: 10.1038/mp.2017.105] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 02/20/2017] [Accepted: 04/04/2017] [Indexed: 12/25/2022]
Abstract
Genetic variations and adverse environmental events in utero or shortly after birth can lead to abnormal brain development and increased risk of schizophrenia. γ-Aminobutyric acid (GABA), the major inhibitory neurotransmitter in the mammalian brain, plays a vital role in normal brain development. GABA synthesis is controlled by enzymes derived from two glutamic acid decarboxylase (GAD) genes, GAD1 and GAD2, both of which produce transcript isoforms. While the full-length GAD1 transcript (GAD67) has been implicated in the neuropathology of schizophrenia, the transcript structure of GAD1 in the human brain has not been fully characterized. In this study, with the use of RNA sequencing and PCR technologies, we report the discovery of 10 novel transcripts of GAD1 in the human brain. Expression levels of four novel GAD1 transcripts (8A, 8B, I80 and I86) showed a lifespan trajectory expression pattern that is anticorrelated with the expression of the full-length GAD1 transcript. In addition, methylation levels of two CpG loci within the putative GAD1 promoter were significantly associated with the schizophrenia-risk SNP rs3749034 and with the expression of GAD25 in dorsolateral prefrontal cortex (DLPFC). Moreover, schizophrenia patients who had completed suicide and/or were positive for nicotine exposure had significantly higher full-length GAD1 expression in the DLPFC. Alternative splicing of GAD1 and epigenetic state appear to play roles in the developmental profile of GAD1 expression and may contribute to GABA dysfunction in the PFC and hippocampus of patients with schizophrenia.
Collapse
Affiliation(s)
- Ran Tao
- The Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA
| | - Kasey N. Davis
- The Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA,Laboratory for Neurodevelopment, Department of Anatomy, Howard University College of Medicine, Washington D.C., USA
| | - Chao Li
- The Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA
| | - Joo Heon Shin
- The Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA
| | - Yuan Gao
- The Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA
| | - Andrew E. Jaffe
- The Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA,Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Marjorie C. Gondré-Lewis
- Laboratory for Neurodevelopment, Department of Anatomy, Howard University College of Medicine, Washington D.C., USA
| | - Daniel R. Weinberger
- The Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA,Department of Psychiatry and Behavior Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, USA,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,McKusick Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Joel E. Kleinman
- The Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA,Department of Psychiatry and Behavior Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Thomas M. Hyde
- The Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA,Department of Psychiatry and Behavior Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, USA,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Roda E, Bottone MG, Insolia V, Barni S, Bernocchi G. Changes in the cerebellar cytoarchitecture of hibernating hedgehog Erinaceus europaeus L. (Mammalia): an immunocytochemical approach. EUROPEAN ZOOLOGICAL JOURNAL 2017. [DOI: 10.1080/24750263.2017.1380722] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- E. Roda
- Department of Biology and Biotechnology “L. Spallanzani”, Laboratory of Cell Biology and Neurobiology, University of Pavia, Pavia, Italy
- Laboratory of Clinical & Experimental Toxicology and Poison Control Centre and National Toxicology Information Centre, Toxicology Unit, ICS Maugeri Spa Benefit Corporation, IRCCS of Pavia, Pavia, Italy
| | - M. G. Bottone
- Department of Biology and Biotechnology “L. Spallanzani”, Laboratory of Cell Biology and Neurobiology, University of Pavia, Pavia, Italy
| | - V. Insolia
- Department of Biology and Biotechnology “L. Spallanzani”, Laboratory of Cell Biology and Neurobiology, University of Pavia, Pavia, Italy
| | - S. Barni
- Department of Biology and Biotechnology “L. Spallanzani”, Laboratory of Cell Biology and Neurobiology, University of Pavia, Pavia, Italy
| | - G. Bernocchi
- Department of Biology and Biotechnology “L. Spallanzani”, Laboratory of Cell Biology and Neurobiology, University of Pavia, Pavia, Italy
| |
Collapse
|
19
|
Fatemi SH, Folsom TD, Liesch SB, Kneeland RE, Karkhane Yousefi M, Thuras PD. The effects of prenatal H1N1 infection at E16 on FMRP, glutamate, GABA, and reelin signaling systems in developing murine cerebellum. J Neurosci Res 2016; 95:1110-1122. [PMID: 27735078 DOI: 10.1002/jnr.23949] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 08/30/2016] [Accepted: 09/06/2016] [Indexed: 12/28/2022]
Abstract
Prenatal viral infection has been identified as a potential risk factor for the development of neurodevelopmental disorders such as schizophrenia and autism. Additionally, dysfunction in gamma-aminobutyric acid, Reelin, and fragile X mental retardation protein (FMRP)-metabotropic glutamate receptor 5 signaling systems has also been demonstrated in these two disorders. In the current report, we have characterized the developmental profiles of selected markers for these systems in cerebella of mice born to pregnant mice infected with human influenza (H1N1) virus on embryonic day 16 or sham-infected controls using SDS-PAGE and Western blotting techniques and evaluated the presence of abnormalities in the above-mentioned markers during brain development. The cerebellum was selected in light of emerging evidence that it plays roles in learning, memory, and emotional processing-all of which are disrupted in autism and schizophrenia. We identified unique patterns of gene and protein expression at birth (postnatal day 0 [P0]), childhood (P14), adolescence (P35), and young adulthood (P56) in both exposed and control mouse progeny. We also identified significant differences in protein expression for FMRP, very-low-density lipoprotein receptor, and glutamic acid decarboxylase 65 and 67 kDa proteins at specific postnatal time points in cerebella of the offspring of exposed mice. Our results provide evidence of disrupted FMRP, glutamatergic, and Reelin signaling in the exposed mouse offspring that explains the multiple brain abnormalities observed in this animal model. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- S Hossein Fatemi
- Department of Psychiatry, Division of Neuroscience Research, University of Minnesota Medical School, Minneapolis, Minnesota.,Department of Neuroscience, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Timothy D Folsom
- Department of Psychiatry, Division of Neuroscience Research, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Stephanie B Liesch
- Department of Psychiatry, Division of Neuroscience Research, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Rachel E Kneeland
- Department of Psychiatry, Division of Neuroscience Research, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Mahtab Karkhane Yousefi
- Department of Psychiatry, Division of Neuroscience Research, University of Minnesota Medical School, Minneapolis, Minnesota.,Department of Psychiatry and Behavioral Neurosciences, Wayne State University, Detroit, Michigan
| | - Paul D Thuras
- VA Medical Center, Department of Psychiatry, Minneapolis, Minnesota
| |
Collapse
|
20
|
Developmental changes in plasticity, synaptic, glia and connectivity protein levels in rat dorsal hippocampus. Neurobiol Learn Mem 2016; 135:125-138. [PMID: 27523749 DOI: 10.1016/j.nlm.2016.08.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 08/06/2016] [Accepted: 08/10/2016] [Indexed: 11/24/2022]
Abstract
Thus far the identification and functional characterization of the molecular mechanisms underlying synaptic plasticity, learning, and memory have not been particularly dissociated from the contribution of developmental changes. Brain plasticity mechanisms have been largely identified and studied using in vitro systems mainly derived from early developmental ages, yet they are considered to be general plasticity mechanisms underlying functions -such as long-term memory- that occurs in the adult brain. Although it is possible that part of the plasticity mechanisms recruited during development is then re-recruited in plasticity responses in adulthood, systematic investigations about whether and how activity-dependent molecular responses differ over development are sparse. Notably, hippocampal-dependent memories are expressed relatively late in development, and the hippocampus undergoes and extended developmental post-natal structural and functional maturation, suggesting that the molecular mechanisms underlying hippocampal neuroplasticity may actually significantly change over development. Here we quantified the relative basal expression levels of sets of plasticity, synaptic, glia and connectivity proteins in rat dorsal hippocampus, a region that is critical for the formation of long-term explicit memories, at two developmental ages, postnatal day 17 (PN17) and PN24, which correspond to a period of relative functional immaturity and maturity, respectively, and compared them to adult age. We found that the levels of numerous proteins and/or their phosphorylation, known to be critical for synaptic plasticity underlying memory formation, including immediate early genes (IEGs), kinases, transcription factors and AMPA receptor subunits, peak at PN17 when the hippocampus is not yet able to express long-term memory. It remains to be established if these changes result from developmental basal activity or infantile learning. Conversely, among all markers investigated, the phosphorylation of calcium calmodulin kinase II α (CamKII α and of extracellular signal-regulated kinases 2 (ERK-2), and the levels of GluA1 and GluA2 significantly increase from PN17 to PN24 and then remain similar in adulthood, thus representing correlates paralleling long-term memory expression ability.
Collapse
|
21
|
Zhuravleva ZN, Hutsyan SS, Zhuravlev GI. Phenotypic differentiation of neurons in intraocular transplants. Russ J Dev Biol 2016. [DOI: 10.1134/s1062360416030085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
22
|
Davis KN, Tao R, Li C, Gao Y, Gondré-Lewis MC, Lipska BK, Shin JH, Xie B, Ye T, Weinberger DR, Kleinman JE, Hyde TM. GAD2 Alternative Transcripts in the Human Prefrontal Cortex, and in Schizophrenia and Affective Disorders. PLoS One 2016; 11:e0148558. [PMID: 26848839 PMCID: PMC4744057 DOI: 10.1371/journal.pone.0148558] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/19/2016] [Indexed: 01/04/2023] Open
Abstract
Genetic variation and early adverse environmental events work together to increase risk for schizophrenia. γ-aminobutyric acid (GABA), the major inhibitory neurotransmitter in adult mammalian brain, plays a major role in normal brain development, and has been strongly implicated in the pathobiology of schizophrenia. GABA synthesis is controlled by two glutamic acid decarboxylase (GAD) genes, GAD1 and GAD2, both of which produce a number of alternative transcripts. Genetic variants in the GAD1 gene are associated with increased risk for schizophrenia, and reduced expression of its major transcript in the human dorsolateral prefrontal cortex (DLPFC). No consistent changes in GAD2 expression have been found in brains from patients with schizophrenia. In this work, with the use of RNA sequencing and PCR technologies, we confirmed and tracked the expression of an alternative truncated transcript of GAD2 (ENST00000428517) in human control DLPFC homogenates across lifespan besides the well-known full length transcript of GAD2. In addition, using quantitative RT-PCR, expression of GAD2 full length and truncated transcripts were measured in the DLPFC of patients with schizophrenia, bipolar disorder and major depression. The expression of GAD2 full length transcript is decreased in the DLPFC of schizophrenia and bipolar disorder patients, while GAD2 truncated transcript is increased in bipolar disorder patients but decreased in schizophrenia patients. Moreover, the patients with schizophrenia with completed suicide or positive nicotine exposure showed significantly higher expression of GAD2 full length transcript. Alternative transcripts of GAD2 may be important in the growth and development of GABA-synthesizing neurons as well as abnormal GABA signaling in the DLPFC of patients with schizophrenia and affective disorders.
Collapse
Affiliation(s)
- Kasey N. Davis
- Clinical Brain Disorders Branch, Genes, Cognition and Psychosis Program, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, 20892–1385, United States of America
- The Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- Laboratory for Neurodevelopment, Department of Anatomy, Howard University College of Medicine, Washington D.C., 20059, United States of America
| | - Ran Tao
- The Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
| | - Chao Li
- The Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
| | - Yuan Gao
- The Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
| | - Marjorie C. Gondré-Lewis
- Laboratory for Neurodevelopment, Department of Anatomy, Howard University College of Medicine, Washington D.C., 20059, United States of America
| | - Barbara K. Lipska
- Clinical Brain Disorders Branch, Genes, Cognition and Psychosis Program, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, 20892–1385, United States of America
| | - Joo Heon Shin
- The Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
| | - Bin Xie
- The Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
| | - Tianzhang Ye
- The Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
| | - Daniel R. Weinberger
- The Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- Department of Psychiatry and Behavior Sciences, and Neurology, Johns Hopkins University School of Medicine, Johns Hopkins University Medical Campus, Baltimore, Maryland, 21205, United States of America
- Departments of Neuroscience and the Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
| | - Joel E. Kleinman
- The Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
| | - Thomas M. Hyde
- The Lieber Institute for Brain Development, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- Department of Psychiatry and Behavior Sciences, and Neurology, Johns Hopkins University School of Medicine, Johns Hopkins University Medical Campus, Baltimore, Maryland, 21205, United States of America
- * E-mail:
| |
Collapse
|
23
|
Pietrzak M. Adhesive capsulitis: An age related symptom of metabolic syndrome and chronic low-grade inflammation? Med Hypotheses 2016; 88:12-7. [PMID: 26880627 DOI: 10.1016/j.mehy.2016.01.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 11/29/2015] [Accepted: 01/01/2016] [Indexed: 02/08/2023]
Abstract
Adhesive capsulitis (AC) is very poorly understood, particularly it's underlying etiology. Obesity and metabolic syndrome, which are strongly associated with chronic low grade inflammation, are becoming increasingly understood to underlie a raft of morbid states including upper limb pain syndromes, diabetes (DM), cardiovascular disease (CVD), cancer and central nervous system dysfunction and degeneration. Notwithstanding age, two of the strongest established risk factors for AC are DM and CVD. The hypothesis argues that similar to DM and CVD, the inflammation and capsular fibrosis seen in AC is precipitated by metabolic syndrome and chronic low grade inflammation. These pathophysiological mechanisms are highly likely to be perpetuated by upregulation of pro-inflammatory cytokine production, sympathetic dominance of autonomic balance, and neuro-immune activation. The hypothesis predicts and describes how these processes may etiologically underpin and induce each sub-classification of AC. An improved understanding of the etiology of AC may lead to more accurate diagnosis, improved management, treatment outcomes, and reduce or prevent pain, disability and suffering associated with the disease. The paper follows on with a discussion of similarities between the pathophysiology of AC to general systemic inflammatory control mechanisms whereby connective tissue (CT) fibrosis is induced as a storage depot for leukocytes and chronic inflammatory cells. The potential role of hyaluronic acid (HA), the primary component of the extracellular matrix (ECM) and CT, in the pathophysiology of AC is also discussed with potential treatment implications. Lastly, a biochemical link between physical and mental health through the ECM is described and the concept of a periventricular-limbic central driver of CT dysfunction is introduced.
Collapse
Affiliation(s)
- Max Pietrzak
- University of Bath, Claverton Down Road, Bath, North East Somerset, BA2 7AY, United Kingdom.
| |
Collapse
|
24
|
Trifonov S, Yamashita Y, Kase M, Maruyama M, Sugimoto T. Glutamic acid decarboxylase 1 alternative splicing isoforms: characterization, expression and quantification in the mouse brain. BMC Neurosci 2014; 15:114. [PMID: 25322942 PMCID: PMC4295415 DOI: 10.1186/1471-2202-15-114] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 09/25/2014] [Indexed: 11/24/2022] Open
Abstract
Background GABA has important functions in brain plasticity related processes like memory, learning, locomotion and during the development of the nervous system. It is synthesized by the glutamic acid decarboxylase (GAD). There are two isoforms of GAD, GAD1 and GAD2, which are encoded by different genes. During embryonic development the transcription of GAD1 mRNA is regulated by alternative splicing and several alternative transcripts were distinguished in human, mouse and rat. Despite the fact that the structure of GAD1 gene has been extensively studied, knowledge of its exact structural organization, alternative promoter usage and splicing have remained incomplete. Results In the present study we report the identification and characterization of novel GAD1 splicing isoforms (GenBank: KM102984, KM102985) by analyzing genomic and mRNA sequence data using bioinformatics, cloning and sequencing. Ten mRNA isoforms are generated from GAD1 gene locus by the combined actions of utilizing different promoters and alternative splicing of the coding exons. Using RT-PCR we found that GAD1 isoforms share similar pattern of expression in different mouse tissues and are expressed early during development. Quantitative RT-PCR was used to investigate the expression of GAD1 isoforms and GAD2 in olfactory bulb, cortex, medial and lateral striatum, hippocampus and cerebellum of adult mouse. Olfactory bulb showed the highest expression of GAD1 transcripts. Isoforms 1/2 are the most abundant forms. Their expression is significantly higher in the lateral compared to the medial striatum. Isoforms 3/4, 5/6, 7/8 and 9/10 are barely detectable in all investigated regions except of the high expression in olfactory bulb. When comparing GAD1 expression with GAD2 we found that Isoforms 1/2 are the predominant isoforms. In situ hybridization confirmed the predominant expression of Isoforms 7/8 and 9/10 in the olfactory bulb and revealed their weak expression in hippocampus, cerebellum and some other areas known to express GAD1. Conclusions Generation of ten splicing isoforms of GAD1 was described including two so far uncharacterized transcripts. GAD1 splicing isoforms producing the shorter, enzymatically inactive GAD25 protein are expressed at very low level in adult mouse brain except in the olfactory bulb that is associated with neurogenesis and synaptic plasticity even during adulthood. Electronic supplementary material The online version of this article (doi:10.1186/1471-2202-15-114) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | - Tetsuo Sugimoto
- Department of Anatomy and Brain Science, Kansai Medical University, Hirakata, Osaka 573-1010, Japan.
| |
Collapse
|
25
|
Jaenisch N, Popp A, Guenther M, Schnabel J, Witte OW, Frahm C. Pro-apoptotic function of GABA-related transcripts following stroke. Neurobiol Dis 2014; 70:237-44. [DOI: 10.1016/j.nbd.2014.06.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 05/28/2014] [Accepted: 06/22/2014] [Indexed: 01/17/2023] Open
|
26
|
Benitez SG, Castro AE, Patterson SI, Muñoz EM, Seltzer AM. Hypoxic preconditioning differentially affects GABAergic and glutamatergic neuronal cells in the injured cerebellum of the neonatal rat. PLoS One 2014; 9:e102056. [PMID: 25032984 PMCID: PMC4102512 DOI: 10.1371/journal.pone.0102056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 06/13/2014] [Indexed: 12/13/2022] Open
Abstract
In this study we examined cerebellar alterations in a neonatal rat model of hypoxic-ischemic brain injury with or without hypoxic preconditioning (Pc). Between postnatal days 7 and 15, the cerebellum is still undergoing intense cellular proliferation, differentiation and migration, dendritogenesis and synaptogenesis. The expression of glutamate decarboxylase 1 (GAD67) and the differentiation factor NeuroD1 were examined as markers of Purkinje and granule cells, respectively. We applied quantitative immunohistochemistry to sagittal cerebellar slices, and Western blot analysis of whole cerebella obtained from control (C) rats and rats submitted to Pc, hypoxia-ischemia (L) and a combination of both treatments (PcL). We found that either hypoxia-ischemia or Pc perturbed the granule cells in the posterior lobes, affecting their migration and final placement in the internal granular layer. These effects were partially attenuated when the Pc was delivered prior to the hypoxia-ischemia. Interestingly, whole nuclear NeuroD1 levels in Pc animals were comparable to those in the C rats. However, a subset of Purkinje cells that were severely affected by the hypoxic-ischemic insult—showing signs of neuronal distress at the levels of the nucleus, cytoplasm and dendritic arborization—were not protected by Pc. A monoclonal antibody specific for GAD67 revealed a three-band pattern in cytoplasmic extracts from whole P15 cerebella. A ∼110 kDa band, interpreted as a potential homodimer of a truncated form of GAD67, was reduced in Pc and L groups while its levels were close to the control animals in PcL rats. Additionally we demonstrated differential glial responses depending on the treatment, including astrogliosis in hypoxiated cerebella and a selective effect of hypoxia-ischemia on the vimentin-immunolabeled intermediate filaments of the Bergmann glia. Thus, while both glutamatergic and GABAergic cerebellar neurons are compromised by the hypoxic-ischemic insult, the former are protected by a preconditioning hypoxia while the latter are not.
Collapse
Affiliation(s)
- Sergio G Benitez
- Laboratory of Neurobiology: Chronobiology Section, Institute of Histology and Embryology of Mendoza (IHEM), School of Medicine, National University of Cuyo, Mendoza, National Scientific and Technical Research Council (CONICET), National Agency for Scientific and Technological Promotion (ANPCyT), Mendoza, Argentina
| | - Analía E Castro
- Laboratory of Neurobiology: Chronobiology Section, Institute of Histology and Embryology of Mendoza (IHEM), School of Medicine, National University of Cuyo, Mendoza, National Scientific and Technical Research Council (CONICET), National Agency for Scientific and Technological Promotion (ANPCyT), Mendoza, Argentina
| | - Sean I Patterson
- Traumatic and Toxic Lesions in the Nervous System Section, Institute of Histology and Embryology of Mendoza (IHEM), School of Medicine, National University of Cuyo, Mendoza, National Scientific and Technical Research Council (CONICET), National Agency for Scientific and Technological Promotion (ANPCyT), Mendoza, Argentina
| | - Estela M Muñoz
- Laboratory of Neurobiology: Chronobiology Section, Institute of Histology and Embryology of Mendoza (IHEM), School of Medicine, National University of Cuyo, Mendoza, National Scientific and Technical Research Council (CONICET), National Agency for Scientific and Technological Promotion (ANPCyT), Mendoza, Argentina
| | - Alicia M Seltzer
- Neonatal Brain Development Section, Institute of Histology and Embryology of Mendoza (IHEM), School of Medicine, National University of Cuyo, Mendoza, National Scientific and Technical Research Council (CONICET), National Agency for Scientific and Technological Promotion (ANPCyT), Mendoza, Argentina
| |
Collapse
|
27
|
Xu W, Xin C, Lin Q, Ding F, Gong W, Zhou Y, Yu J, Cui P, Hu S. Adolescent mouse takes on an active transcriptomic expression during postnatal cerebral development. GENOMICS PROTEOMICS & BIOINFORMATICS 2014; 12:111-9. [PMID: 24953867 PMCID: PMC4411375 DOI: 10.1016/j.gpb.2014.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 04/02/2014] [Accepted: 04/03/2014] [Indexed: 12/15/2022]
Abstract
Postnatal cerebral development is a complicated biological process precisely controlled by multiple genes. To understand the molecular mechanism of cerebral development, we compared dynamics of mouse cerebrum transcriptome through three developmental stages using high-throughput RNA-seq technique. Three libraries were generated from the mouse cerebrum at infancy, adolescence and adulthood, respectively. Consequently, 44,557,729 (infancy), 59,257,530 (adolescence) and 72,729,636 (adulthood) reads were produced, which were assembled into 15,344, 16,048 and 15,775 genes, respectively. We found that the overall gene expression level increased from infancy to adolescence and decreased later on upon reaching adulthood. The adolescence cerebrum has the most active gene expression, with expression of a large number of regulatory genes up-regulated and some crucial pathways activated. Transcription factor (TF) analysis suggested the similar dynamics as expression profiling, especially those TFs functioning in neurogenesis differentiation, oligodendrocyte lineage determination and circadian rhythm regulation. Moreover, our data revealed a drastic increase in myelin basic protein (MBP)-coding gene expression in adolescence and adulthood, suggesting that the brain myelin may be generated since mouse adolescence. In addition, differential gene expression analysis indicated the activation of rhythmic pathway, suggesting the function of rhythmic movement since adolescence; Furthermore, during infancy and adolescence periods, gene expression related to axon repulsion and attraction showed the opposite trends, indicating that axon repulsion was activated after birth, while axon attraction might be activated at the embryonic stage and declined during the postnatal development. Our results from the present study may shed light on the molecular mechanism underlying the postnatal development of the mammalian cerebrum.
Collapse
Affiliation(s)
- Wei Xu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Chengqi Xin
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Qiang Lin
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Feng Ding
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Wei Gong
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuanyuan Zhou
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jun Yu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Peng Cui
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Songnian Hu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
28
|
Deidda G, Bozarth IF, Cancedda L. Modulation of GABAergic transmission in development and neurodevelopmental disorders: investigating physiology and pathology to gain therapeutic perspectives. Front Cell Neurosci 2014; 8:119. [PMID: 24904277 PMCID: PMC4033255 DOI: 10.3389/fncel.2014.00119] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/14/2014] [Indexed: 01/30/2023] Open
Abstract
During mammalian ontogenesis, the neurotransmitter GABA is a fundamental regulator of neuronal networks. In neuronal development, GABAergic signaling regulates neural proliferation, migration, differentiation, and neuronal-network wiring. In the adult, GABA orchestrates the activity of different neuronal cell-types largely interconnected, by powerfully modulating synaptic activity. GABA exerts these functions by binding to chloride-permeable ionotropic GABAA receptors and metabotropic GABAB receptors. According to its functional importance during development, GABA is implicated in a number of neurodevelopmental disorders such as autism, Fragile X, Rett syndrome, Down syndrome, schizophrenia, Tourette's syndrome and neurofibromatosis. The strength and polarity of GABAergic transmission is continuously modulated during physiological, but also pathological conditions. For GABAergic transmission through GABAA receptors, strength regulation is achieved by different mechanisms such as modulation of GABAA receptors themselves, variation of intracellular chloride concentration, and alteration in GABA metabolism. In the never-ending effort to find possible treatments for GABA-related neurological diseases, of great importance would be modulating GABAergic transmission in a safe and possibly physiological way, without the dangers of either silencing network activity or causing epileptic seizures. In this review, we will discuss the different ways to modulate GABAergic transmission normally at work both during physiological and pathological conditions. Our aim is to highlight new research perspectives for therapeutic treatments that reinstate natural and physiological brain functions in neuro-pathological conditions.
Collapse
Affiliation(s)
- Gabriele Deidda
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia Genova, Italy
| | - Ignacio F Bozarth
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia Genova, Italy
| | - Laura Cancedda
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia Genova, Italy
| |
Collapse
|
29
|
Boulland JL, Chaudhry FA. Ontogenetic changes in the distribution of the vesicular GABA transporter VGAT correlate with the excitation/inhibition shift of GABA action. Neurochem Int 2012; 61:506-16. [PMID: 22490609 DOI: 10.1016/j.neuint.2012.03.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 03/19/2012] [Accepted: 03/20/2012] [Indexed: 10/28/2022]
Abstract
GABA is the major inhibitory neurotransmitter in the adult CNS and is among others involved in the synchronization of large neuronal networks. During development, GABA acts as a morphogenetic factor and has transient excitatory actions in many brain regions. One distinct protein, the vesicular GABA transporter (VGAT), has been identified accumulating GABA into presynaptic vesicles prior to its exocytotic release. The function of VGAT and its distribution is well defined in the adult, but its contribution to the transient excitatory action at putative GABAergic nerve terminals in the immature brain and its potential roles in putative glutamatergic nerve terminals remain elusive. We have studied VGAT expression in the brain from late embryonic stages through several postnatal stages until adulthood. Quantitative immunoblotting and immunolabeling of tissue sections at the light microscope and the electron microscope levels show an abrupt augmentation in VGAT staining in the cerebral cortex during the first three postnatal weeks, resembling the increase in other proteins involved in GABA synthesis and recycling in the same time frame - such as GAD65, GAD67, GAT1 (Slc6a1) and SN1 (Slc38a3) - and coincides with the synaptogenetic spurt. Dynamic changes in the expression of VGAT are seen in many cellular populations and in several layers in different brain regions. However, mossy fiber terminals (MFT) elude staining for VGAT. We also demonstrate that VGAT(+) nerve terminals undergo a developmental reorganization so that from targeting primarily the dendrites of the principal neurons in several brain regions in the immature brain, they target the soma of the same cells in the adult. This shift in the targeted subcellular compartment coincides with the conversion of the chloride gradient across neuronal membranes and suggests that it may be important for the shift of GABA action from excitation to inhibition and for the establishment of the potent synchronization of neuronal networks.
Collapse
|
30
|
Piccolini V, Cerri S, Romanelli E, Bernocchi G. Interactions of neurotransmitter systems during postnatal development of the rat hippocampal formation: Effects of cisplatin. Exp Neurol 2012; 234:239-52. [DOI: 10.1016/j.expneurol.2011.12.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 12/21/2011] [Accepted: 12/25/2011] [Indexed: 01/14/2023]
|
31
|
de Azevedo-Pereira RL, Lima APCA, Rodrigues DDC, Rondinelli E, Medei EH, Goldenberg RC, de Carvalho ACCC, Mendez-Otero R. Cysteine proteases in differentiation of embryonic stem cells into neural cells. Stem Cells Dev 2011; 20:1859-72. [PMID: 21417836 DOI: 10.1089/scd.2010.0186] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Glycosylated mouse cystatin C (mCysC), an endogenous inhibitor of cysteine cathepsin proteases (CP), has been suggested as a cofactor of β-FGF to induce the differentiation of mouse embryonic stem cells into neural progenitor cells (NPCs). To investigate the possible role of CP in neural differentiation, we treated embryoid bodies (EBs) with (i) E64, an inhibitor of papain-like CP and of calpains, (ii) an inhibitor of cathepsin L (iCatL), (iii) an inhibitor of calpains (iCalp), or (iv) cystatins, and their ability to differentiate into neural cells was assessed. We show that the inhibition of CP induces a significant increase in Pax6 expression in EBs, leading to an increase in the number of nestin-positive cells after 3 days. Fourteen days after E64 treatment, we observed increased numbers of β-III-tubulin-positive cells, showing greater percentage of immature neurons, and this feature persisted up to 24 days. At this point, we encountered higher numbers of neurons with inward Na(+) current compared with untreated EBs. Further, we show that mCysC and iCatL, but not unglycosylated egg white cystatin or iCalp, increased the numbers of NPCs. In contrast to E64 and iCatL, mCysC did not inhibit CP in EBs and its neural-inducing activity required β-FGF. We propose that the inhibition of CP induces the differentiation of mouse embryonic stem cells into NPCs and neurons through a mechanism that is distinct from CysC-induced neural differentiation.
Collapse
|
32
|
Abstract
The neurotransmitter GABA (γ-aminobutyric acid), acting via inotropic GABA(A) and metabotropic GABA(B) receptors, plays an essential role in a variety of distinct neuronal processes, including regulation of neuronal excitability, determination of temporal aspects of spike trains, control of the size and propagation of neuronal assemblies, generation of oscillatory activity, and neuronal plasticity. Although the developmental switch between excitatory and inhibitory GABA(A) receptor-mediated responses is widely appreciated, the fact that the postnatal maturation of the GABAergic system lasts until late adolescence is not so persuasively promoted. This review summarizes recent knowledge of the maturation of various aspects of the GABAergic systems, like functional expression of GABA synthesizing/degrading enzymes and transporters, density of GABAergic synapses, GABAergic projection patterns, GABA receptor subunit composition, and properties of GABAergic interneurons, with an emphasis on the late developmental alterations. In addition, some aspects of the development of mental capabilities during adolescence and their relation the delayed maturation of the GABAergic system are presented.
Collapse
Affiliation(s)
- Werner Kilb
- Institute of Physiology and Pathophysiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
33
|
Downregulation of hippocampal GABA after hypoxia-induced seizures in neonatal rats. Neurochem Res 2011; 36:2409-16. [PMID: 21833845 DOI: 10.1007/s11064-011-0565-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2010] [Revised: 05/28/2011] [Accepted: 07/27/2011] [Indexed: 02/04/2023]
Abstract
This study aims to determine the expression of Gamma-aminobutyric acid (GABA) following hypoxia in neonatal rats and explore how it may increase susceptibility to epilepsy later in life. A modified model of neonatal hypoxia-induced epileptic susceptibility was simulated by 17 min of hypoxia (5% O(2) and 95% N(2)) in postnatal day (P) 10 rats. Hippocampal glutamate decarboxylase (GAD) and parvalbumin (PV) during the development with or without hypoxia were examined using immunohistochemistry. No detectable neuronal loss was observed in the hippocampus either immediately or 14 days after hypoxia. During the development GAD- and PV-immunoreactivity increased substantially during P 11-13 and reached mature expression in the control rats, and decreased significantly at different time points except for a transient increase during P 11-13 in the hypoxic groups. Our study indicates that downregulation of hippocampal GABA after hypoxia-induced seizures in neonatal rats may contribute to higher epileptic susceptibility in later life.
Collapse
|
34
|
Hyde TM, Lipska BK, Ali T, Mathew SV, Law AJ, Metitiri OE, Straub RE, Ye T, Colantuoni C, Herman MM, Bigelow LB, Weinberger DR, Kleinman JE. Expression of GABA signaling molecules KCC2, NKCC1, and GAD1 in cortical development and schizophrenia. J Neurosci 2011; 31:11088-95. [PMID: 21795557 PMCID: PMC3758549 DOI: 10.1523/jneurosci.1234-11.2011] [Citation(s) in RCA: 230] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Revised: 05/19/2011] [Accepted: 05/30/2011] [Indexed: 01/18/2023] Open
Abstract
GABA signaling molecules are critical for both human brain development and the pathophysiology of schizophrenia. We examined the expression of transcripts derived from three genes related to GABA signaling [GAD1 (GAD67 and GAD25), SLC12A2 (NKCC1), and SLC12A5 (KCC2)] in the prefrontal cortex (PFC) and hippocampal formation of a large cohort of nonpsychiatric control human brains (n = 240) across the lifespan (from fetal week 14 to 80 years) and in patients with schizophrenia (n = 30-31), using quantitative RT-PCR. We also examined whether a schizophrenia risk-associated promoter SNP in GAD1 (rs3749034) is related to expression of these transcripts. Our studies revealed that development and maturation of both the PFC and hippocampal formation are characterized by progressive switches in expression from GAD25 to GAD67 and from NKCC1 to KCC2. Previous studies have demonstrated that the former leads to GABA synthesis, and the latter leads to switching from excitatory to inhibitory neurotransmission. In the hippocampal formation, GAD25/GAD67 and NKCC1/KCC2 ratios are increased in patients with schizophrenia, reflecting a potentially immature GABA physiology. Remarkably, GAD25/GAD67 and NKCC1/KCC2 expression ratios are associated with rs3749034 genotype, with risk alleles again predicting a relatively less mature pattern. These findings suggest that abnormalities in GABA signaling critical to brain development contribute to genetic risk for schizophrenia.
Collapse
Affiliation(s)
- Thomas M Hyde
- Section on Neuropathology, Clinical Brain Disorders Branch, Genes, Cognition and Psychosis Program, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Compensatory network alterations upon onset of epilepsy in synapsin triple knock-out mice. Neuroscience 2011; 189:108-22. [PMID: 21621590 DOI: 10.1016/j.neuroscience.2011.05.030] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Revised: 04/16/2011] [Accepted: 05/11/2011] [Indexed: 11/21/2022]
Abstract
Adult synapsin triple-knockout mice exhibit epilepsy that manifests as generalized tonic-clonic seizures. Because in vitro recordings have shown a reduction in quantal release from inhibitory neurons, an inherent excitation-inhibition imbalance has been hypothesized as the direct culprit for epilepsy in these mice. We critically assessed this hypothesis by examining neurotransmission during the emergence of epilepsy. Using long-term video and telemetric EEG monitoring we found that synapsin triple-knockout mice exhibit an abrupt transition during early adulthood from a seizure-free presymptomatic latent state to a consistent symptomatic state of sensory-induced seizures. Electrophysiological recordings showed that during the latent period larger field responses could be elicited in slices from mutant mice. However, only after the transition to a symptomatic state in the adult mice did evoked epileptiform activity become prevalent. This state was characterized by resistance to the epileptiform-promoting effects of 4-aminopyridine, by marked hypersensitivity to blockage of GABAA receptors, and by the emergence of unresponsiveness to NMDA receptor antagonism, all of which were not observed during the latent period. Importantly, enhancement in inhibitory transmission was associated with upregulation of GAD67 expression without affecting the number of inhibitory neurons in the same brain areas where epileptiform activity was recorded. We therefore suggest that while deletion of the synapsins initially increases cortical network activity, this enhanced excitability is insufficient to elicit seizures. Rather, compensatory epileptogenic mechanisms are activated during the latent period that lead to an additional almost-balanced enhancement of both the excitatory and inhibitory components of the network, finally culminating in the emergence of epilepsy.
Collapse
|
36
|
Fish KN, Sweet RA, Lewis DA. Differential distribution of proteins regulating GABA synthesis and reuptake in axon boutons of subpopulations of cortical interneurons. ACTA ACUST UNITED AC 2011; 21:2450-60. [PMID: 21422269 DOI: 10.1093/cercor/bhr007] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Subclasses of γ-aminobutyric acid (GABA) interneurons differentially influence cortical network activity. The contribution of differences in GABA synthesis and reuptake in axon boutons to cell type-specific functions is unknown. GABA is synthesized within boutons by glutamic acid decarboxylase 65 (GAD65) and GAD67, while GAT1 is responsible for GABA reuptake. Using an imaging methodology capable of determining the colocalization frequency of different immunocytochemical labels in the same bouton and the quantification of the fluorescence intensity of each label in these same structures, we assessed the bouton levels of GAD65, GAD67, and GAT1 in parvalbumin-expressing chandelier (PV(ch)) and basket (PV(b)) neurons and cannabinoid 1 receptor-expressing basket (CB1r(b)) neurons in the monkey prefrontal cortex. We show that PV(ch) boutons almost exclusively contained GAD67, relative to GAD65, whereas CB1r(b) boutons contained mostly GAD65. In contrast, both GAD65 and GAD67 were easily detected in PV(b) boutons. Furthermore, in comparison with PV(ch) boutons, CB1r(b) boutons expressed low to undetectable levels of GAT1. Our findings provide a new basis for the distinctive functional roles of these perisomatic-innervating interneurons in cortical circuits. In addition, they strongly suggest that altered levels of GAD67 or GAD65, as seen in some psychiatric diseases, would have cell type-specific consequences on the modulation of GABA neurotransmission.
Collapse
Affiliation(s)
- Kenneth N Fish
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | | | | |
Collapse
|
37
|
Infragranular gene expression disturbances in the prefrontal cortex in schizophrenia: signature of altered neural development? Neurobiol Dis 2009; 37:738-46. [PMID: 20034564 DOI: 10.1016/j.nbd.2009.12.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 12/10/2009] [Accepted: 12/14/2009] [Indexed: 12/11/2022] Open
Abstract
The development of the human neocortex gives rise to a complex cytoarchitecture, grouping together cells with similar structure, connectivity and function. As a result, the six neocortical laminae show distinct molecular content. In schizophrenia, many anatomical and neurochemical changes appear to be restricted to a subset of lamina and/or cell types. In this study, we hypothesized that supragranular (SG; laminae II-III) and infragranular layers (IG; laminae V-VI) of area 46 in the human prefrontal cortex will show distinct and specific transcriptome alterations between subjects with schizophrenia and matched controls. To enhance sample homogeneity, we compared the gene expression patterns of the SG and IG layers of 8 matched middle-aged male subjects with schizophrenia to 8 pairwise matched controls using two replicate DNA microarrays for each sample. The study revealed strong disease-related laminar expression differences between the SG and IG layers. Expression changes were dominated by an overall underexpression of the IG-enriched genes in the schizophrenia subjects compared to normal control subjects. Furthermore, using a diagnosis-blind, unsupervised clustering of the control-derived SG or IG-enriched transcripts, the IG-enriched markers segregated the subjects with schizophrenia from the matched controls with a high degree of confidence. Importantly, multiple members of the semaphorin gene family reported altered gene expression, suggesting that the IG gene expression disturbances in subjects with schizophrenia may be a result of altered cortical development and disrupted brain connectivity.
Collapse
|