1
|
Wadhwa M, Sall JW, Chinn GA. Neonatal Diazepam Exposure Decreases Dendritic Arborization and Spine Density of Cortical Pyramidal Neurons in Rats. J Neurosurg Anesthesiol 2025; 37:225-231. [PMID: 38973590 DOI: 10.1097/ana.0000000000000979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/06/2024] [Indexed: 07/09/2024]
Abstract
OBJECTIVE Benzodiazepines are extensively utilized in pediatric anesthesia and critical care for their anxiolytic and sedative properties. However, preclinical studies indicate that neonatal exposure to GABAergic drugs, including benzodiazepines, leads to long-term cognitive deficits, potentially mediated by altered GABAergic signaling during brain development. This preclinical study investigated the impact of early-life diazepam exposure on cortical neuronal morphology, specifically exploring dendritic arborization and spine density, crucial factors in synaptogenesis. METHODS Male and female Sprague Dawley rat pups were exposed to a single neonatal dose of diazepam (30 mg/kg) or vehicle on postnatal day (PND) 7. Golgi-Cox staining was used to assess cortical pyramidal neuron development at 4 developmental stages: neonatal (PND8), infantile (PND15), juvenile (PND30), and adolescence (PND42). Animals were randomized equally to 4 groups: male-vehicle, male-diazepam, female-vehicle, and female-diazepam. Neuronal morphology was evaluated after reconstruction in neurolucida, and dendritic spine density was analyzed through high-power photomicrographs using ImageJ. RESULTS Diazepam exposure resulted in decreased dendritic complexity in both sexes, with reduced arborization and spine density observed in cortical pyramidal neurons. Significant differences were found at each developmental stage, indicating a persistent impact. Dendritic length increased with age but was attenuated by diazepam exposure. Branching length analysis revealed decreased complexity after diazepam treatment. Spine density at PND42 was significantly reduced in both apical and basal dendrites after diazepam exposure. CONCLUSIONS Neonatal diazepam exposure adversely affected cortical pyramidal neuron development, leading to persistent alterations in dendritic arborization and spine density. These structural changes suggest potential risks associated with early-life diazepam exposure. Further research is needed to unravel the functional consequences of these anatomic alterations.
Collapse
Affiliation(s)
- Meetu Wadhwa
- Department of Anesthesia and Perioperative Care, University of California, San Francisco (UCSF), San Francisco, CA
| | | | | |
Collapse
|
2
|
Tzakis N, Ethier-Gagnon M, Epp T, Holahan MR. Assessment of cFos labeling in the hippocampus and anterior cingulate cortex following recent and remote re-exposure to an unreinforced open field in preadolescent and postadolescent rats. Behav Brain Res 2025; 476:115284. [PMID: 39393683 DOI: 10.1016/j.bbr.2024.115284] [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: 08/13/2024] [Revised: 09/26/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024]
Abstract
Spatial tasks are often goal-directed or reward-facilitated confounding the assessment of "pure" recent and remote spatial memories. The current work re-exposed preadolescent and postadolescent male rats to a non-reinforced, free exploration task to investigate cFos patterns within the hippocampus and anterior cingulate cortex (ACC) associated with recent and remote periods. Male rats were exposed to an open field task for one, 30 min session on postnatal day (P) 20, 25, or 50 and re-exposed for 30 min at either a recent (24 hours) or remote (3 weeks) timepoint. Distance traveled in the open field was measured as well as cFos labeling. In the P20 age group, there was elevated exploration at the 24-hour and 3-week tests compared to training and compared to the other age groups. In the hippocampus CA1, cFos levels were higher after the remote test than the recent test in the P20 group but higher after the recent test than remote test in the P25 and P50 groups. cFos labeling in the ACC was higher in all remote-tested groups compared to the recent-tested groups across all ages. In the P20, the 24-hour test was associated with less CA1 activity than the other age groups supporting the hypothesis that the hippocampus is not fully developed at this time point. In the P20 group, the remote representation of this task did not seem to be complete as there continued to be CA1 activity along with ACC activity following the remote test associated with elevated exploration. These results indicate the utility of unreinforced spatial navigation tasks for exploring systems consolidation processes over the lifespan and show that a fully developed hippocampus is required for optimal systems consolidation.
Collapse
Affiliation(s)
- Nikolaos Tzakis
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada
| | | | - Tanisse Epp
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada
| | - Matthew R Holahan
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada.
| |
Collapse
|
3
|
Lalzad A, Wong F, Schneider M. Neuroinflammation in the Rat Brain After Exposure to Diagnostic Ultrasound. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:961-968. [PMID: 38685265 DOI: 10.1016/j.ultrasmedbio.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/19/2023] [Accepted: 02/11/2024] [Indexed: 05/02/2024]
Abstract
OBJECTIVE To date there have been no studies exploring the potential for neuroinflammation as an intracranial bio-effect associated with diagnostic ultrasound during neonatal cranial scans in a mammalian in vivo model. The study described here was aimed at investigating the effects of B-mode and Doppler mode ultrasound on inflammation in the rat brain. METHODS Twelve Wistar rats (7-9 wk old) were divided into a control group and an ultrasound-exposed group (n = 6/group). A craniotomy was performed, followed by 10 min of B-mode and spectral Doppler interrogation of the middle cerebral artery. The control group was subjected to sham treatment, with the transducer held stationary over the craniotomy site, but the ultrasound machine switched off. Animals were euthanized 48 h after exposure, and the brains formalin fixed for immunohistochemical analysis using allograft inflammatory factor 1 (IBA-1) and glial fibrillary acidic protein (GFAP) as markers of microglia and astrocytes, respectively. The numbers of IBA-1- and GFAP-immunoreactive cells were manually counted and expressed as areal density (cells/mm2). Results were analyzed using Student's unpaired t-test and one-way repeated-measures analysis of variance. RESULTS The ultrasound-exposed brain exhibited significant increases in IBA-1 and GFAP immunoreactive cell density in all regions of B-mode and Doppler mode exposure compared with the control group (p < 0.001). CONCLUSION Ten minutes of B-mode and Doppler mode ultrasound may induce neuroinflammatory changes in the rat brain. This suggests that exposure of brain tissue to current diagnostic ultrasound intensities may not be completely without risk.
Collapse
Affiliation(s)
- Assema Lalzad
- Department of Medical Imaging and Radiation Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia; Department of Medical Imaging, Cabrini Hospital, Malvern, Victoria, Australia
| | - Flora Wong
- Monash Newborn, Monash Medical Centre, Clayton, Victoria, Australia; The Ritchie Centre, Hudson's Institute of Medical Research, Melbourne, Victoria; Department of Pediatrics, Monash University, Clayton, Victoria, Australia
| | - Michal Schneider
- Department of Medical Imaging and Radiation Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
4
|
Radzishevsky I, Odeh M, Bodner O, Zubedat S, Shaulov L, Litvak M, Esaki K, Yoshikawa T, Agranovich B, Li WH, Radzishevsky A, Gottlieb E, Avital A, Wolosker H. Impairment of serine transport across the blood-brain barrier by deletion of Slc38a5 causes developmental delay and motor dysfunction. Proc Natl Acad Sci U S A 2023; 120:e2302780120. [PMID: 37812701 PMCID: PMC10589673 DOI: 10.1073/pnas.2302780120] [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: 02/17/2023] [Accepted: 09/08/2023] [Indexed: 10/11/2023] Open
Abstract
Brain L-serine is critical for neurodevelopment and is thought to be synthesized solely from glucose. In contrast, we found that the influx of L-serine across the blood-brain barrier (BBB) is essential for brain development. We identified the endothelial Slc38a5, previously thought to be a glutamine transporter, as an L-serine transporter expressed at the BBB in early postnatal life. Young Slc38a5 knockout (KO) mice exhibit developmental alterations and a decrease in brain L-serine and D-serine, without changes in serum or liver amino acids. Slc38a5-KO brains exhibit accumulation of neurotoxic deoxysphingolipids, synaptic and mitochondrial abnormalities, and decreased neurogenesis at the dentate gyrus. Slc38a5-KO pups exhibit motor impairments that are affected by the administration of L-serine at concentrations that replenish the serine pool in the brain. Our results highlight a critical role of Slc38a5 in supplying L-serine via the BBB for proper brain development.
Collapse
Affiliation(s)
- Inna Radzishevsky
- Department of Biochemistry, B. Rappaport Faculty of Medicine, Technion-Israel Institue of Technology, Haifa3109601, Israel
| | - Maali Odeh
- Department of Biochemistry, B. Rappaport Faculty of Medicine, Technion-Israel Institue of Technology, Haifa3109601, Israel
| | - Oded Bodner
- Department of Biochemistry, B. Rappaport Faculty of Medicine, Technion-Israel Institue of Technology, Haifa3109601, Israel
| | - Salman Zubedat
- Department of Occupational Therapy, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa3498838, Israel
| | - Lihi Shaulov
- Electron Microscopy Unit, B. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa3109601, Israel
| | - Maxim Litvak
- Department of Biochemistry, B. Rappaport Faculty of Medicine, Technion-Israel Institue of Technology, Haifa3109601, Israel
| | - Kayoko Esaki
- Department of Biotechnology and Life Sciences, Faculty of Biotechnology and Life Sciences, Sojo University, Kumamoto860-0082, Japan
| | - Takeo Yoshikawa
- Laboratory of Molecular Psychiatry, RIKEN Center for Brain Science, Saitama351-0198, Japan
| | - Bella Agranovich
- Laura and Isaac Perlmutter Metabolomics Center, B. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa3109601, Israel
| | - Wen-Hong Li
- Department of Cell Biology and Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX75390-9039
| | | | - Eyal Gottlieb
- Technion-Integrated Cancer Center, B. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa3109601, Israel
| | - Avi Avital
- Department of Occupational Therapy, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa3498838, Israel
| | - Herman Wolosker
- Department of Biochemistry, B. Rappaport Faculty of Medicine, Technion-Israel Institue of Technology, Haifa3109601, Israel
- Laura and Isaac Perlmutter Metabolomics Center, B. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa3109601, Israel
| |
Collapse
|
5
|
Puzio M, Moreton N, Sullivan M, Scaife C, Glennon JC, O'Connor JJ. An Electrophysiological and Proteomic Analysis of the Effects of the Superoxide Dismutase Mimetic, MnTMPyP, on Synaptic Signalling Post-Ischemia in Isolated Rat Hippocampal Slices. Antioxidants (Basel) 2023; 12:antiox12040792. [PMID: 37107167 PMCID: PMC10135248 DOI: 10.3390/antiox12040792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Metabolic stress and the increased production of reactive oxygen species (ROS) are two main contributors to neuronal damage and synaptic plasticity in acute ischemic stroke. The superoxide scavenger MnTMPyP has been previously reported to have a neuroprotective effect in organotypic hippocampal slices and to modulate synaptic transmission after in vitro hypoxia and oxygen-glucose deprivation (OGD). However, the mechanisms involved in the effect of this scavenger remain elusive. In this study, two concentrations of MnTMPyP were evaluated on synaptic transmission during ischemia and post-ischemic synaptic potentiation. The complex molecular changes supporting cellular adaptation to metabolic stress, and how these are modulated by MnTMPyP, were also investigated. Electrophysiological data showed that MnTMPyP causes a decrease in baseline synaptic transmission and impairment of synaptic potentiation. Proteomic analysis performed on MnTMPyP and hypoxia-treated tissue indicated an impairment in vesicular trafficking mechanisms, including reduced expression of Hsp90 and actin signalling. Alterations of vesicular trafficking may lead to reduced probability of neurotransmitter release and AMPA receptor activity, resulting in the observed modulatory effect of MnTMPyP. In OGD, protein enrichment analysis highlighted impairments in cell proliferation and differentiation, such as TGFβ1 and CDKN1B signalling, in addition to downregulation of mitochondrial dysfunction and an increased expression of CAMKII. Taken together, our results may indicate modulation of neuronal sensitivity to the ischemic insult, and a complex role for MnTMPyP in synaptic transmission and plasticity, potentially providing molecular insights into the mechanisms mediating the effects of MnTMPyP during ischemia.
Collapse
Affiliation(s)
- Martina Puzio
- UCD School of Biomolecular & Biomedical Science, University College Dublin, Dublin 4, Ireland
- Mass Spectrometry Core Facility, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Niamh Moreton
- UCD School of Biomolecular & Biomedical Science, University College Dublin, Dublin 4, Ireland
- Mass Spectrometry Core Facility, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Mairéad Sullivan
- Mass Spectrometry Core Facility, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
- UCD School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Caitriona Scaife
- Mass Spectrometry Core Facility, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Jeffrey C Glennon
- Mass Spectrometry Core Facility, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
- UCD School of Medicine, University College Dublin, Dublin 4, Ireland
| | - John J O'Connor
- UCD School of Biomolecular & Biomedical Science, University College Dublin, Dublin 4, Ireland
- Mass Spectrometry Core Facility, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| |
Collapse
|
6
|
Howes OD, Shatalina E. Integrating the Neurodevelopmental and Dopamine Hypotheses of Schizophrenia and the Role of Cortical Excitation-Inhibition Balance. Biol Psychiatry 2022; 92:501-513. [PMID: 36008036 DOI: 10.1016/j.biopsych.2022.06.017] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/16/2022] [Accepted: 06/04/2022] [Indexed: 12/23/2022]
Abstract
The neurodevelopmental and dopamine hypotheses are leading theories of the pathoetiology of schizophrenia, but they were developed in isolation. However, since they were originally proposed, there have been considerable advances in our understanding of the normal neurodevelopmental refinement of synapses and cortical excitation-inhibition (E/I) balance, as well as preclinical findings on the interrelationship between cortical and subcortical systems and new in vivo imaging and induced pluripotent stem cell evidence for lower synaptic density markers in patients with schizophrenia. Genetic advances show that schizophrenia is associated with variants linked to genes affecting GABA (gamma-aminobutyric acid) and glutamatergic signaling as well as neurodevelopmental processes. Moreover, in vivo studies on the effects of stress, particularly during later development, show that it leads to synaptic elimination. We review these lines of evidence as well as in vivo evidence for altered cortical E/I balance and dopaminergic dysfunction in schizophrenia. We discuss mechanisms through which frontal cortex circuitry may regulate striatal dopamine and consider how frontal E/I imbalance may cause dopaminergic dysregulation to result in psychotic symptoms. This integrated neurodevelopmental and dopamine hypothesis suggests that overpruning of synapses, potentially including glutamatergic inputs onto frontal cortical interneurons, disrupts the E/I balance and thus underlies cognitive and negative symptoms. It could also lead to disinhibition of excitatory projections from the frontal cortex and possibly other regions that regulate mesostriatal dopamine neurons, resulting in dopamine dysregulation and psychotic symptoms. Together, this explains a number of aspects of the epidemiology and clinical presentation of schizophrenia and identifies new targets for treatment and prevention.
Collapse
Affiliation(s)
- Oliver D Howes
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Hammersmith Hospital, Imperial College London, United Kingdom; Department of Psychosis, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom.
| | - Ekaterina Shatalina
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Hammersmith Hospital, Imperial College London, United Kingdom
| |
Collapse
|
7
|
Moreton N, Puzio M, O’Connor JJ. The effects of the superoxide dismutase mimetic, MnTMPyP, post hypoxia and oxygen glucose deprivation, in isolated rat hippocampal slices. Brain Res Bull 2022; 190:105-115. [DOI: 10.1016/j.brainresbull.2022.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/17/2022] [Accepted: 09/27/2022] [Indexed: 11/02/2022]
|
8
|
Lippert RN, Brüning JC. Maternal Metabolic Programming of the Developing Central Nervous System: Unified Pathways to Metabolic and Psychiatric Disorders. Biol Psychiatry 2022; 91:898-906. [PMID: 34330407 DOI: 10.1016/j.biopsych.2021.06.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/17/2022]
Abstract
The perinatal period presents a critical time in offspring development where environmental insults can have damaging impacts on the future health of the offspring. This can lead to sustained alterations in offspring development, metabolism, and predisposition to both metabolic and psychiatric diseases. The central nervous system is one of the most sensitive targets in response to maternal obesity and/or type 2 diabetes mellitus. While many of the effects of obesity on brain function in adults are known, we are only now beginning to understand the multitude of changes that occur in the brain during development on exposure to maternal overnutrition. Specifically, given recent links between maternal metabolic state and onset of neurodevelopmental diseases, the specific changes that are occurring in the offspring are even more relevant for the study of disease onset. It is therefore critical to understand the developmental effects of maternal obesity and/or type 2 diabetes mellitus and further to define the underlying cellular and molecular changes in the fetal brain. This review focuses on the current advancements in the study of maternal programming of brain development with particular emphasis on brain connectivity, specific regional effects, newly studied peripheral contributors, and key windows of interventions where maternal bodyweight and food intake may drive the most detrimental effects on the brain and associated metabolic and behavioral consequences.
Collapse
Affiliation(s)
- Rachel N Lippert
- German Institute of Human Nutrition Potsdam Rehbrücke, Potsdam, Germany; German Center for Diabetes Research, Neuherberg, Germany; Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Jens C Brüning
- German Center for Diabetes Research, Neuherberg, Germany; Max Planck Institute for Metabolism Research, Cologne, Germany; Policlinic for Endocrinology, Diabetes and Preventive Medicine, University Hospital Cologne, Cologne, Germany.
| |
Collapse
|
9
|
Alrousan G, Hassan A, Pillai AA, Atrooz F, Salim S. Early Life Sleep Deprivation and Brain Development: Insights From Human and Animal Studies. Front Neurosci 2022; 16:833786. [PMID: 35592259 PMCID: PMC9111737 DOI: 10.3389/fnins.2022.833786] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 03/25/2022] [Indexed: 11/13/2022] Open
Abstract
Adequate sleep especially during developmental stages of life, is considered essential for normal brain development and believed to play an important role in promoting healthy cognitive and psychosocial development, while persistent sleep disturbances and/or sleep deprivation during early life are believed to trigger many mental ailments such as anxiety disorders, depression, and cognitive impairment. Initially it was suggested that adverse mental health conditions adversely affect sleep, however, it is now accepted that this association is bidirectional. In fact, sleep disturbances are listed as a symptom of many mental health disorders. Of special interest is the association between early life sleep deprivation and its negative mental health outcomes. Studies have linked persistent early life sleep deprivation with later life behavioral and cognitive disturbances. Neurobiological underpinnings responsible for the negative outcomes of early life sleep deprivation are not understood. This is a significant barrier for early therapeutic and/or behavioral intervention, which can be feasible only if biological underpinnings are well-understood. Animal studies have provided useful insights in this area. This article focusses on the knowledge gained from the research conducted in the area of early life sleep deprivation, brain development, and behavioral function studies.
Collapse
Affiliation(s)
- Ghalya Alrousan
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Arham Hassan
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Aditya Anilkumar Pillai
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Fatin Atrooz
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Samina Salim
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| |
Collapse
|
10
|
Super-resolved 3D-STED microscopy identifies a layer-specific increase in excitatory synapses in the hippocampal CA1 region of Neuroligin-3 KO mice. Biochem Biophys Res Commun 2021; 582:144-149. [PMID: 34715405 DOI: 10.1016/j.bbrc.2021.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/02/2021] [Indexed: 12/18/2022]
Abstract
The chemical synapse is one type of cell-adhesion system that transmits information from a neuron to another neuron in the complex neuronal network in the brain. Synaptic transmission is the rate-limiting step during the information processing in the neuronal network and its plasticity is involved in cognitive functions. Thus, morphological and electrophysiological analyses of synapses are of particular importance in neuroscience research. In the current study, we applied super-resolved three-dimensional stimulated emission depletion (3D-STED) microscopy for the morphological analyses of synapses. This approach allowed us to estimate the precise number of excitatory and inhibitory synapses in the mouse hippocampal tissue. We discovered a region-specific increase in excitatory synapses in a model mouse of autism spectrum disorder, Neuroligin-3 KO, with this method. This type of analysis will open a new field in developmental neuroscience in the future.
Collapse
|
11
|
Parker KN, Donovan MH, Smith K, Noble-Haeusslein LJ. Traumatic Injury to the Developing Brain: Emerging Relationship to Early Life Stress. Front Neurol 2021; 12:708800. [PMID: 34484104 PMCID: PMC8416304 DOI: 10.3389/fneur.2021.708800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/22/2021] [Indexed: 12/01/2022] Open
Abstract
Despite the high incidence of brain injuries in children, we have yet to fully understand the unique vulnerability of a young brain to an injury and key determinants of long-term recovery. Here we consider how early life stress may influence recovery after an early age brain injury. Studies of early life stress alone reveal persistent structural and functional impairments at adulthood. We consider the interacting pathologies imposed by early life stress and subsequent brain injuries during early brain development as well as at adulthood. This review outlines how early life stress primes the immune cells of the brain and periphery to elicit a heightened response to injury. While the focus of this review is on early age traumatic brain injuries, there is also a consideration of preclinical models of neonatal hypoxia and stroke, as each further speaks to the vulnerability of the brain and reinforces those characteristics that are common across each of these injuries. Lastly, we identify a common mechanistic trend; namely, early life stress worsens outcomes independent of its temporal proximity to a brain injury.
Collapse
Affiliation(s)
- Kaila N. Parker
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Michael H. Donovan
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Kylee Smith
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Linda J. Noble-Haeusslein
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
12
|
Traetta ME, Codagnone MG, Uccelli NA, Ramos AJ, Zárate S, Reinés A. Hippocampal neurons isolated from rats subjected to the valproic acid model mimic in vivo synaptic pattern: evidence of neuronal priming during early development in autism spectrum disorders. Mol Autism 2021; 12:23. [PMID: 33676530 PMCID: PMC7937248 DOI: 10.1186/s13229-021-00428-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Autism spectrum disorders (ASD) are synaptopathies characterized by area-specific synaptic alterations and neuroinflammation. Structural and adhesive features of hippocampal synapses have been described in the valproic acid (VPA) model. However, neuronal and microglial contribution to hippocampal synaptic pattern and its time-course of appearance is still unknown. METHODS Male pups born from pregnant rats injected at embryonic day 10.5 with VPA (450 mg/kg, i.p.) or saline (control) were used. Maturation, exploratory activity and social interaction were assessed as autistic-like traits. Synaptic, cell adhesion and microglial markers were evaluated in the CA3 hippocampal region at postnatal day (PND) 3 and 35. Primary cultures of hippocampal neurons from control and VPA animals were used to study synaptic features and glutamate-induced structural remodeling. Basal and stimuli-mediated reactivity was assessed on microglia primary cultures isolated from control and VPA animals. RESULTS At PND3, before VPA behavioral deficits were evident, synaptophysin immunoreactivity and the balance between the neuronal cell adhesion molecule (NCAM) and its polysialylated form (PSA-NCAM) were preserved in the hippocampus of VPA animals along with the absence of microgliosis. At PND35, concomitantly with the establishment of behavioral deficits, the hippocampus of VPA rats showed fewer excitatory synapses and increased NCAM/PSA-NCAM balance without microgliosis. Hippocampal neurons from VPA animals in culture exhibited a preserved synaptic puncta number at the beginning of the synaptogenic period in vitro but showed fewer excitatory synapses as well as increased NCAM/PSA-NCAM balance and resistance to glutamate-induced structural synaptic remodeling after active synaptogenesis. Microglial cells isolated from VPA animals and cultured in the absence of neurons showed similar basal and stimuli-induced reactivity to the control group. Results indicate that in the absence of glia, hippocampal neurons from VPA animals mirrored the in vivo synaptic pattern and suggest that while neurons are primed during the prenatal period, hippocampal microglia are not intrinsically altered. CONCLUSIONS Our study suggests microglial role is not determinant for developing neuronal alterations or counteracting neuronal outcome in the hippocampus and highlights the crucial role of hippocampal neurons and structural plasticity in the establishment of the synaptic alterations in the VPA rat model.
Collapse
Affiliation(s)
- Marianela Evelyn Traetta
- Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis” (IBCN), Facultad de Medicina, CONICET - Universidad de Buenos Aires, Calle Paraguay 2155 3er piso, 1121 Ciudad de Buenos Aires, Argentina
- Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Martín Gabriel Codagnone
- Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis” (IBCN), Facultad de Medicina, CONICET - Universidad de Buenos Aires, Calle Paraguay 2155 3er piso, 1121 Ciudad de Buenos Aires, Argentina
- Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nonthué Alejandra Uccelli
- Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis” (IBCN), Facultad de Medicina, CONICET - Universidad de Buenos Aires, Calle Paraguay 2155 3er piso, 1121 Ciudad de Buenos Aires, Argentina
| | - Alberto Javier Ramos
- Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis” (IBCN), Facultad de Medicina, CONICET - Universidad de Buenos Aires, Calle Paraguay 2155 3er piso, 1121 Ciudad de Buenos Aires, Argentina
- Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sandra Zárate
- Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Analía Reinés
- Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis” (IBCN), Facultad de Medicina, CONICET - Universidad de Buenos Aires, Calle Paraguay 2155 3er piso, 1121 Ciudad de Buenos Aires, Argentina
- Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
13
|
Philippot G, Hellsten SV, Viberg H, Fredriksson R. Evaluation of the dentate gyrus in adult mice exposed to acetaminophen (paracetamol) on postnatal day 10. Int J Dev Neurosci 2020; 81:91-97. [PMID: 33222217 DOI: 10.1002/jdn.10079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/16/2020] [Accepted: 11/16/2020] [Indexed: 11/09/2022] Open
Abstract
Acetaminophen (AAP; or paracetamol) is a widely used nonprescription drug with antipyretic and analgesic properties. Alarmingly, there is an increasing body of evidence showing that developmental exposure to AAP is associated with adverse behavioural outcomes later in life. We have previously shown that relevant doses of AAP in 10-day-old mice affected memory, learning and locomotor activity in the adult animals. Interestingly, the neurons of the dentate gyrus (DG) have a relatively late time of origin as they are generated during the first two weeks of postnatal life in rodents. Since the generation of these cells, which are important for memory processing, coincides with our AAP exposure, we aim to investigate if the cytoarchitecture of the DG is affected by postnatal day 10 AAP treatment. In addition, we investigate if markers for differentiation and migration in the hippocampus were affected by the same treatment. We did not observe any visual effects in adult DG cytoarchitecture, nor any changes of markers for differentiation/migration in the hippocampus in 24 hr after exposure. Even though a large effect size was estimated on adult DG thickness following AAP exposure, the estimated 95% CIs around the differences of the means reveal no significant effect. Hence, larger sample sizes are warranted to clarify if neonatal AAP exposure affects adult DG thickness in mice.
Collapse
Affiliation(s)
- Gaëtan Philippot
- Department of Pharmaceutical Biosciences, Molecular Neuropharmacology, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Sofie V Hellsten
- Department of Pharmaceutical Biosciences, Molecular Neuropharmacology, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Henrik Viberg
- Department of Organismal Biology, Environmental Toxicology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
| | - Robert Fredriksson
- Department of Pharmaceutical Biosciences, Molecular Neuropharmacology, Biomedical Center, Uppsala University, Uppsala, Sweden
| |
Collapse
|
14
|
Pollitt SL, Myers KR, Yoo J, Zheng JQ. LIM and SH3 protein 1 localizes to the leading edge of protruding lamellipodia and regulates axon development. Mol Biol Cell 2020; 31:2718-2732. [PMID: 32997597 PMCID: PMC7927181 DOI: 10.1091/mbc.e20-06-0366] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The actin cytoskeleton drives cell motility and is essential for neuronal development and function. LIM and SH3 protein 1 (LASP1) is a unique actin-binding protein that is expressed in a wide range of cells including neurons, but its roles in cellular motility and neuronal development are not well understood. We report that LASP1 is expressed in rat hippocampus early in development, and this expression is maintained through adulthood. High-resolution imaging reveals that LASP1 is selectively concentrated at the leading edge of lamellipodia in migrating cells and axonal growth cones. This local enrichment of LASP1 is dynamically associated with the protrusive activity of lamellipodia, depends on the barbed ends of actin filaments, and requires both the LIM domain and the nebulin repeats of LASP1. Knockdown of LASP1 in cultured rat hippocampal neurons results in a substantial reduction in axonal outgrowth and arborization. Finally, loss of the Drosophila homologue Lasp from a subset of commissural neurons in the developing ventral nerve cord produces defasciculated axon bundles that do not reach their targets. Together, our data support a novel role for LASP1 in actin-based lamellipodial protrusion and establish LASP1 as a positive regulator of both in vitro and in vivo axon development.
Collapse
Affiliation(s)
| | | | - Jin Yoo
- Emory College, Emory University, Atlanta, GA 30322
| | - James Q Zheng
- Department of Cell Biology and.,Department of Neurology and Center for Neurodegenerative Diseases, Emory University School of Medicine, and
| |
Collapse
|
15
|
Cizeron M, Qiu Z, Koniaris B, Gokhale R, Komiyama NH, Fransén E, Grant SGN. A brainwide atlas of synapses across the mouse life span. Science 2020; 369:270-275. [PMID: 32527927 PMCID: PMC7115813 DOI: 10.1126/science.aba3163] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 05/19/2020] [Indexed: 01/03/2023]
Abstract
Synapses connect neurons together to form the circuits of the brain, and their molecular composition controls innate and learned behavior. We analyzed the molecular and morphological diversity of 5 billion excitatory synapses at single-synapse resolution across the mouse brain from birth to old age. A continuum of changes alters synapse composition in all brain regions across the life span. Expansion in synapse diversity produces differentiation of brain regions until early adulthood, and compositional changes cause dedifferentiation in old age. The spatiotemporal synaptome architecture of the brain potentially accounts for life-span transitions in intellectual ability, memory, and susceptibility to behavioral disorders.
Collapse
Affiliation(s)
- Mélissa Cizeron
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Institut NeuroMyoGène, Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, 69008 Lyon, France
| | - Zhen Qiu
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Babis Koniaris
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- School of Computing, Edinburgh Napier University, Edinburgh EH10 5DT, UK
| | - Ragini Gokhale
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Noboru H Komiyama
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Erik Fransén
- Department of Computational Science and Technology, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, 10044 Stockholm, Sweden
- Science for Life Laboratory, KTH Royal Institute of Technology, SE-171 65 Solna, Sweden
| | - Seth G N Grant
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK.
- Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| |
Collapse
|
16
|
Chou ZZ, Yu GJ, Berger TW. Generation of Granule Cell Dendritic Morphologies by Estimating the Spatial Heterogeneity of Dendritic Branching. Front Comput Neurosci 2020; 14:23. [PMID: 32327990 PMCID: PMC7160759 DOI: 10.3389/fncom.2020.00023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 03/13/2020] [Indexed: 11/13/2022] Open
Abstract
Biological realism of dendritic morphologies is important for simulating electrical stimulation of brain tissue. By adding point process modeling and conditional sampling to existing generation strategies, we provide a novel means of reproducing the nuanced branching behavior that occurs in different layers of granule cell dendritic morphologies. In this study, a heterogeneous Poisson point process was used to simulate branching events. Conditional distributions were then used to select branch angles depending on the orthogonal distance to the somatic plane. The proposed method was compared to an existing generation tool and a control version of the proposed method that used a homogeneous Poisson point process. Morphologies were generated with each method and then compared to a set of digitally reconstructed neurons. The introduction of a conditionally dependent branching rate resulted in the generation of morphologies that more accurately reproduced the emergent properties of dendritic material per layer, Sholl intersections, and proximal passive current flow. Conditional dependence was critically important for the generation of realistic granule cell dendritic morphologies.
Collapse
Affiliation(s)
- Zane Z Chou
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, United States
| | - Gene J Yu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, United States
| | - Theodore W Berger
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
17
|
Short AK, Maras PM, Pham AL, Ivy AS, Baram TZ. Blocking CRH receptors in adults mitigates age-related memory impairments provoked by early-life adversity. Neuropsychopharmacology 2020; 45:515-523. [PMID: 31698409 PMCID: PMC6969076 DOI: 10.1038/s41386-019-0562-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/26/2019] [Accepted: 10/28/2019] [Indexed: 01/06/2023]
Abstract
In humans, early-life adversity is associated with impairments in learning and memory that may emerge later in life. In rodent models, early-life adversity directly impacts hippocampal neuron structure and connectivity with progressive deficits in long-term potentiation and spatial memory function. Previous work has demonstrated that augmented release and actions of the stress-activated neuropeptide, CRH, contribute to the deleterious effects of early-life adversity on hippocampal dendritic arborization, synapse number and memory-function. Early-life adversity increases hippocampal CRH expression, and blocking hippocampal CRH receptor type-1 (CRHR1) immediately following early-life adversity prevented the consequent memory and LTP defects. Here, we tested if blocking CRHR1 in young adults ameliorates early-life adversity-provoked memory deficits later in life. A weeklong course of a CRHR1 antagonist in 2-month-old male rats prevented early-life adversity-induced deficits in object recognition memory that emerged by 12 months of age. Surprisingly, whereas the intervention did not mitigate early-life adversity-induced spatial memory losses at 4 and 8 months, it restored hippocampus-dependent location memory in 12-month-old rats that experienced early-life adversity. Neither early-life adversity nor CRHR1 blockade in the adult influenced anxiety- or depression-related behaviors. Altogether, these findings suggest that cognitive deficits attributable to adversity during early-life-sensitive periods are at least partially amenable to interventions later in life.
Collapse
Affiliation(s)
- Annabel K Short
- Department of Anatomy and Neurobiology, University of California- Irvine, Irvine, CA, USA.
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA.
| | - Pamela M Maras
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Aidan L Pham
- Department of Anatomy and Neurobiology, University of California- Irvine, Irvine, CA, USA
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
| | - Autumn S Ivy
- Department of Anatomy and Neurobiology, University of California- Irvine, Irvine, CA, USA
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Department of Anatomy and Neurobiology, University of California- Irvine, Irvine, CA, USA
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
- Department of Neurology, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
18
|
Atrooz F, Salim S. Sleep deprivation, oxidative stress and inflammation. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 119:309-336. [PMID: 31997771 DOI: 10.1016/bs.apcsb.2019.03.001] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Adequate sleep is essential for normal brain function, especially during early life developmental stages as postnatal brain maturation occurs during the critical period of childhood and adolescence. Therefore, sleep disturbance and/or deficit during this period can have detrimental consequences. Many epidemiological and clinical studies have linked early life sleep disturbance with occurrence of later life behavioral and cognitive impairments. Role of oxidative stress and inflammation has been implicated in sleep deprivation-related impairments. This review article presents a detailed description of the current state of the literature on the subject.
Collapse
Affiliation(s)
- Fatin Atrooz
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Samina Salim
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| |
Collapse
|
19
|
Raber J, Arzy S, Bertolus JB, Depue B, Haas HE, Hofmann SG, Kangas M, Kensinger E, Lowry CA, Marusak HA, Minnier J, Mouly AM, Mühlberger A, Norrholm SD, Peltonen K, Pinna G, Rabinak C, Shiban Y, Soreq H, van der Kooij MA, Lowe L, Weingast LT, Yamashita P, Boutros SW. Current understanding of fear learning and memory in humans and animal models and the value of a linguistic approach for analyzing fear learning and memory in humans. Neurosci Biobehav Rev 2019; 105:136-177. [PMID: 30970272 DOI: 10.1016/j.neubiorev.2019.03.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 01/30/2019] [Accepted: 03/18/2019] [Indexed: 01/04/2023]
Abstract
Fear is an emotion that serves as a driving factor in how organisms move through the world. In this review, we discuss the current understandings of the subjective experience of fear and the related biological processes involved in fear learning and memory. We first provide an overview of fear learning and memory in humans and animal models, encompassing the neurocircuitry and molecular mechanisms, the influence of genetic and environmental factors, and how fear learning paradigms have contributed to treatments for fear-related disorders, such as posttraumatic stress disorder. Current treatments as well as novel strategies, such as targeting the perisynaptic environment and use of virtual reality, are addressed. We review research on the subjective experience of fear and the role of autobiographical memory in fear-related disorders. We also discuss the gaps in our understanding of fear learning and memory, and the degree of consensus in the field. Lastly, the development of linguistic tools for assessments and treatment of fear learning and memory disorders is discussed.
Collapse
Affiliation(s)
- Jacob Raber
- Department of Behavioral Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR, USA; Departments of Neurology and Radiation Medicine, and Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR, USA.
| | - Shahar Arzy
- Department of Medical Neurobiology, Hebrew University, Jerusalem 91904, Israel
| | | | - Brendan Depue
- Departments of Psychological and Brain Sciences and Anatomical Sciences and Neurobiology, University of Louisville, Louisville, KY, USA
| | - Haley E Haas
- Department of Psychiatry and Behavioral Science, Emory University School of Medicine, Atlanta, GA, USA
| | - Stefan G Hofmann
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA
| | - Maria Kangas
- Department of Psychology, Macquarie University, Sydney, Australia
| | | | - Christopher A Lowry
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Hilary A Marusak
- Department of Pharmacy Practice, Wayne State University, Detroit, MI, USA
| | - Jessica Minnier
- School of Public Health, Oregon Health & Science University, Portland, OR, USA
| | - Anne-Marie Mouly
- Lyon Neuroscience Research Center, CNRS-UMR 5292, INSERM U1028, Université Lyon, Lyon, France
| | - Andreas Mühlberger
- Department of Psychology (Clinical Psychology and Psychotherapy), University of Regensburg, Regensburg, Germany; PFH - Private University of Applied Sciences, Department of Psychology (Clinical Psychology and Psychotherapy Research), Göttingen, Germany
| | - Seth Davin Norrholm
- Department of Psychiatry and Behavioral Science, Emory University School of Medicine, Atlanta, GA, USA
| | - Kirsi Peltonen
- Faculty of Social Sciences/Psychology, Tampere University, Tampere, Finland
| | - Graziano Pinna
- The Psychiatric Institute, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Christine Rabinak
- Department of Pharmacy Practice, Wayne State University, Detroit, MI, USA
| | - Youssef Shiban
- Department of Psychology (Clinical Psychology and Psychotherapy), University of Regensburg, Regensburg, Germany; PFH - Private University of Applied Sciences, Department of Psychology (Clinical Psychology and Psychotherapy Research), Göttingen, Germany
| | - Hermona Soreq
- Department of Biological Chemistry, Edmond and Lily Safra Center of Brain Science and The Institute of Life Sciences, Hebrew University, Jerusalem 91904, Israel
| | - Michael A van der Kooij
- Translational Psychiatry, Department of Psychiatry and Psychotherapy, Universitatsmedizin der Johannes Guttenberg University Medical Center, Mainz, Germany
| | | | - Leah T Weingast
- Department of Psychiatry and Behavioral Science, Emory University School of Medicine, Atlanta, GA, USA
| | - Paula Yamashita
- School of Public Health, Oregon Health & Science University, Portland, OR, USA
| | - Sydney Weber Boutros
- Department of Behavioral Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
20
|
Differential expression of SV2A in hippocampal glutamatergic and GABAergic terminals during postnatal development. Brain Res 2019; 1715:73-83. [PMID: 30905653 DOI: 10.1016/j.brainres.2019.03.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/08/2019] [Accepted: 03/20/2019] [Indexed: 01/15/2023]
Abstract
The function of synaptic vesicle protein 2A (SV2A) has not been clearly identified, although it has an essential role in normal neurotransmission. Changes in SV2A expression have been linked to several diseases that could implicate an imbalance between excitation and inhibition, such as epilepsy. Although it is known that SV2A expression is necessary for survival, SV2A expression and its relationship with γ-aminobutyric acid (GABA) and glutamate neurotransmitter systems along development has not been addressed. This report follows SV2A expression levels in the rat hippocampus and their association with glutamatergic and GABAergic terminals along postnatal development. Total SV2A expression was assessed by real time PCR and western blot, while immunofluorescence was used to identify SV2A protein in the different hippocampal layers and its co-localization with GABA or glutamate vesicular transporters. SV2A was dynamically regulated along development and its association with GABA or glutamate transporters varied in the different hippocampal layers. In the principal cells layers (granular and pyramidal), SV2A protein was preferentially localized to GABAergic terminals, while in the hilus and stratum lucidum SV2A was associated mainly to glutamatergic terminals. Although SV2A was ubiquitously expressed in the entire hippocampus, it established a differential association with excitatory or inhibitory terminals, which could contribute to the maturation of excitatory/inhibitory balance.
Collapse
|
21
|
Preferential Targeting of Lateral Entorhinal Inputs onto Newly Integrated Granule Cells. J Neurosci 2018; 38:5843-5853. [PMID: 29793975 DOI: 10.1523/jneurosci.1737-17.2018] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 04/26/2018] [Accepted: 05/17/2018] [Indexed: 12/20/2022] Open
Abstract
Mature dentate granule cells in the hippocampus receive input from the entorhinal cortex via the perforant path in precisely arranged lamina, with medial entorhinal axons innervating the middle molecular layer and lateral entorhinal cortex axons innervating the outer molecular layer. Although vastly outnumbered by mature granule cells, adult-generated newborn granule cells play a unique role in hippocampal function, which has largely been attributed to their enhanced excitability and plasticity (Schmidt-Hieber et al., 2004; Ge et al., 2007). Inputs from the medial and lateral entorhinal cortex carry different informational content. Thus, the distribution of inputs onto newly integrated granule cells will affect their function in the circuit. Using retroviral labeling in combination with selective optogenetic activation of medial or lateral entorhinal inputs, we examined the functional innervation and synaptic maturation of newly generated dentate granule cells in the mouse hippocampus. Our results indicate that lateral entorhinal inputs provide the majority of functional innervation of newly integrated granule cells at 21 d postmitosis. Despite preferential functional targeting, the dendritic spine density of immature granule cells was similar in the outer and middle molecular layers, which we speculate could reflect an unequal distribution of shaft synapses. However, chronic blockade of neurotransmitter release of medial entorhinal axons with tetanus toxin disrupted normal synapse development of both medial and lateral entorhinal inputs. Our results support a role for preferential lateral perforant path input onto newly generated neurons in mediating pattern separation, but also indicate that medial perforant path input is necessary for normal synaptic development.SIGNIFICANCE STATEMENT The formation of episodic memories involves the integration of contextual and spatial information. Newly integrated neurons in the dentate gyrus of the hippocampus play a critical role in this process, despite constituting only a minor fraction of the total number of granule cells. Here we demonstrate that these neurons preferentially receive information thought to convey the context of an experience. Each newly integrated granule cell plays this unique role for ∼1 month before reaching maturity.
Collapse
|
22
|
Jolly S, Bazargani N, Quiroga AC, Pringle NP, Attwell D, Richardson WD, Li H. G protein-coupled receptor 37-like 1 modulates astrocyte glutamate transporters and neuronal NMDA receptors and is neuroprotective in ischemia. Glia 2017; 66:47-61. [PMID: 28795439 PMCID: PMC5724489 DOI: 10.1002/glia.23198] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 07/05/2017] [Accepted: 07/13/2017] [Indexed: 12/21/2022]
Abstract
We show that the G protein‐coupled receptor GPR37‐like 1 (GPR37L1) is expressed in most astrocytes and some oligodendrocyte precursors in the mouse central nervous system. This contrasts with GPR37, which is mainly in mature oligodendrocytes. Comparison of wild type and Gpr37l1–/– mice showed that loss of GPR37L1 did not affect the input resistance or resting potential of astrocytes or neurons in the hippocampus. However, GPR37L1‐mediated signalling inhibited astrocyte glutamate transporters and – surprisingly, given its lack of expression in neurons – reduced neuronal NMDA receptor (NMDAR) activity during prolonged activation of the receptors as occurs in ischemia. This effect on NMDAR signalling was not mediated by a change in the release of D‐serine or TNF‐α, two astrocyte‐derived agents known to modulate NMDAR function. After middle cerebral artery occlusion, Gpr37l1 expression was increased around the lesion. Neuronal death was increased by ∼40% in Gpr37l1–/– brain compared to wild type in an in vitro model of ischemia. Thus, GPR37L1 protects neurons during ischemia, presumably by modulating extracellular glutamate concentration and NMDAR activation.
Collapse
Affiliation(s)
- Sarah Jolly
- Wolfson Institute for Biomedical Research, University College London, London, WC1E 6BT, United Kingdom
| | - Narges Bazargani
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, United Kingdom
| | - Alejandra C Quiroga
- Wolfson Institute for Biomedical Research, University College London, London, WC1E 6BT, United Kingdom
| | - Nigel P Pringle
- Wolfson Institute for Biomedical Research, University College London, London, WC1E 6BT, United Kingdom
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, United Kingdom
| | - William D Richardson
- Wolfson Institute for Biomedical Research, University College London, London, WC1E 6BT, United Kingdom
| | - Huiliang Li
- Wolfson Institute for Biomedical Research, University College London, London, WC1E 6BT, United Kingdom
| |
Collapse
|
23
|
Enduring Neural and Behavioral Effects of Early Life Adversity in Infancy: Consequences of Maternal Abuse and Neglect, Trauma and Fear. Curr Behav Neurosci Rep 2017. [DOI: 10.1007/s40473-017-0112-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
24
|
Lauterborn JC, Kramár EA, Rice JD, Babayan AH, Cox CD, Karsten CA, Gall CM, Lynch G. Cofilin Activation Is Temporally Associated with the Cessation of Growth in the Developing Hippocampus. Cereb Cortex 2017; 27:2640-2651. [PMID: 27073215 PMCID: PMC5964364 DOI: 10.1093/cercor/bhw088] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Dendritic extension and synaptogenesis proceed at high rates in rat hippocampus during early postnatal life but markedly slow during the third week of development. The reasons for the latter, fundamental event are poorly understood. Here, we report that levels of phosphorylated (inactive) cofilin, an actin depolymerizing factor, decrease by 90% from postnatal days (pnds) 10 to 21. During the same period, levels of total and phosphorylated Arp2, which nucleates actin branches, increase. A search for elements that could explain the switch from inactive to active cofilin identified reductions in β1 integrin, TrkB, and LIM domain kinase 2b, upstream proteins that promote cofilin phosphorylation. Moreover, levels of slingshot 3, which dephosphorylates cofilin, increase during the period in which growth slows. Consistent with the cofilin results, in situ phalloidin labeling of F-actin demonstrated that spines and dendrites contained high levels of dynamic actin filaments during Week 2, but these fell dramatically by pnd 21. The results suggest that the change from inactive to constitutively active cofilin leads to a loss of dynamic actin filaments needed for process extension and thus the termination of spine formation and synaptogenesis. The relevance of these events to the emergence of memory-related synaptic plasticity is described.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Christine M. Gall
- Department of Anatomy and Neurobiology
- Department of Neurobiology and Behavior
| | - Gary Lynch
- Department of Anatomy and Neurobiology
- Department of Psychiatry and Human Behavior, University of California at Irvine, Irvine, CA 92697, USA
| |
Collapse
|
25
|
Neonatal blockade of GABA‐A receptors alters behavioral and physiological phenotypes in adult mice. Int J Dev Neurosci 2017; 57:62-71. [DOI: 10.1016/j.ijdevneu.2017.01.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 12/11/2016] [Accepted: 01/16/2017] [Indexed: 11/21/2022] Open
|
26
|
Boulanger Bertolus J, Mouly AM, Sullivan RM. Ecologically relevant neurobehavioral assessment of the development of threat learning. Learn Mem 2016; 23:556-66. [PMID: 27634146 PMCID: PMC5026204 DOI: 10.1101/lm.042218.116] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 06/02/2016] [Indexed: 11/24/2022]
Abstract
As altricial infants gradually transition to adults, their proximate environment changes. In three short weeks, pups transition from a small world with the caregiver and siblings to a complex milieu rich in dangers as their environment expands. Such contrasting environments require different learning abilities and lead to distinct responses throughout development. Here, we will review some of the learned fear conditioned responses to threats in rats during their ontogeny, including behavioral and physiological measures that permit the assessment of learning and its supporting neurobiology from infancy through adulthood. In adulthood, odor-shock conditioning produces robust fear learning to the odor that depends upon the amygdala and related circuitry. Paradoxically, this conditioning in young pups fails to support fear learning and supports approach learning to the odor previously paired with shock. This approach learning is mediated by the infant attachment network that does not include the amygdala. During the age range when pups transition from the infant to the adult circuit (10-15 d old), pups have access to both networks: odor-shock conditioning in maternal presence uses the attachment circuit but the adult amygdala-dependent circuit when alone. However, throughout development (as young as 5 d old) the attachment associated learning can be overridden and amygdala-dependent fear learning supported, if the mother expresses fear in the presence of the pup. This social modulation of the fear permits the expression of defense reactions in life threatening situations informed by the caregiver but prevents the learning of the caregiver itself as a threat.
Collapse
Affiliation(s)
| | - Anne-Marie Mouly
- Lyon Neuroscience Research Center, INSERM U1028; CNRS UMR5292; University Lyon1, Lyon, France
| | - Regina M Sullivan
- Emotional Brain Institute, Nathan Kline Institute, Child and Adolescent Psychiatry, New York University School of Medicine, New York, New York 10010, USA
| |
Collapse
|
27
|
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: 41] [Impact Index Per Article: 4.6] [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
|
28
|
Ontogeny of memory: An update on 40 years of work on infantile amnesia. Behav Brain Res 2016; 298:4-14. [DOI: 10.1016/j.bbr.2015.07.030] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 07/08/2015] [Accepted: 07/08/2015] [Indexed: 01/01/2023]
|
29
|
Abstract
Due to a high incidence of traumatic brain injury (TBI) in children and adolescents, age-specific studies are necessary to fully understand the long-term consequences of injuries to the immature brain. Preclinical and translational research can help elucidate the vulnerabilities of the developing brain to insult, and provide model systems to formulate and evaluate potential treatments aimed at minimizing the adverse effects of TBI. Several experimental TBI models have therefore been scaled down from adult rodents for use in juvenile animals. The following chapter discusses these adapted models for pediatric TBI, and the importance of age equivalence across species during model development and interpretation. Many neurodevelopmental processes are ongoing throughout childhood and adolescence, such that neuropathological mechanisms secondary to a brain insult, including oxidative stress, metabolic dysfunction and inflammation, may be influenced by the age at the time of insult. The long-term evaluation of clinically relevant functional outcomes is imperative to better understand the persistence and evolution of behavioral deficits over time after injury to the developing brain. Strategies to modify or protect against the chronic consequences of pediatric TBI, by supporting the trajectory of normal brain development, have the potential to improve quality of life for brain-injured children.
Collapse
Affiliation(s)
- Bridgette D Semple
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Jaclyn Carlson
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Linda J Noble-Haeusslein
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA.
- Department of Physical Therapy and Rehabilitation Science, University of California School of Medicine, 513 Parnassus Ave., HSE 814, San Francisco, CA, 94143, USA.
| |
Collapse
|
30
|
Different expressions of large-conductance Ca2+-activated K+ channels in the mouse renal cortex and hippocampus during postnatal development. Appl Immunohistochem Mol Morphol 2015; 23:146-52. [PMID: 25390352 DOI: 10.1097/pai.0000000000000006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Large-conductance Ca(+)-activated K(+) (BKCa) channels are widely distributed in a variety of cells and play a pivotal and specific role in many pathophysiological conditions. However, the function of BK(Ca) channels in the kidney cortex and hippocampus during the postnatal development has not received attention. In this study, to elucidate the role of BK(Ca) channels during the development, it is essential to establish the location and quantitation of expression of BK(Ca). The expressions of BK(Ca) were detected in the kidney and hippocampus on postnatal days (P) 1, 3, 5, 7, 14, 21, 28, and 49 by immunohistochemical and Western blot analysis. Our results showed that expressions of BK(Ca) channels were found in tubules and corpuscles at all time points. The expression was also observed at all developmental stages of the renal corpuscles, such as comma-shaped body, S-shaped body, renal corpuscles of stage III, and renal corpuscles of stage IV. During the development, the expression of BK(Ca) channels was decreased and the most prominent change of BK(Ca) protein level appeared between P14 and P21. In contrast, BK(Ca) channels were expressed in all regions of the hippocampus at every time point with the level increasing during the early development (P1 to P14). The findings of the present study suggest that BKCa channels play an important role during the postnatal development in both the renal cortex and hippocampus.
Collapse
|
31
|
Smith CA, Farmer K, Lee H, Holahan MR, Smith JC. Altered Hippocampal Lipid Profile Following Acute Postnatal Exposure to Di(2-Ethylhexyl) Phthalate in Rats. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2015; 12:13542-59. [PMID: 26516880 PMCID: PMC4627048 DOI: 10.3390/ijerph121013542] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 10/14/2015] [Accepted: 10/20/2015] [Indexed: 12/13/2022]
Abstract
Slight changes in the abundance of certain lipid species in the brain may drastically alter normal neurodevelopment via membrane stability, cell signalling, and cell survival. Previous findings have demonstrated that postnatal exposure to di (2-ethylhexyl) phthalate (DEHP) disrupts normal axonal and neural development in the hippocampus. The goal of the current study was to determine whether postnatal exposure to DEHP alters the lipid profile in the hippocampus during postnatal development. Systemic treatment with 10 mg/kg DEHP during postnatal development led to elevated levels of phosphatidylcholine and sphingomyelin in the hippocampus of female rats. There was no effect of DEHP exposure on the overall abundance of phosphatidylcholine or sphingomyelin in male rats or of lysophosphatidylcholine in male or female rats. Individual analyses of each identified lipid species revealed 10 phosphatidylcholine and six sphingomyelin lipids in DEHP-treated females and a single lysophosphatidylcholine in DEHP-treated males with a two-fold or higher increase in relative abundance. Our results are congruent with previous work that found that postnatal exposure to DEHP had a near-selective detrimental effect on hippocampal development in males but not females. Together, results suggest a neuroprotective effect of these elevated lipid species in females.
Collapse
Affiliation(s)
- Catherine A Smith
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, 325 LSRB, Ottawa ON, K1S 5B6, Canada.
| | - Kyle Farmer
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, 325 LSRB, Ottawa ON, K1S 5B6, Canada.
| | - Hyunmin Lee
- Department of Chemistry, Carleton University, Ottawa, 1125 Colonel By Drive, SC-226, Ottawa, ON, K1S5B6, Canada.
| | - Matthew R Holahan
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, 325 LSRB, Ottawa ON, K1S 5B6, Canada.
| | - Jeffrey C Smith
- Department of Chemistry, Carleton University, Ottawa, 1125 Colonel By Drive, SC-226, Ottawa, ON, K1S5B6, Canada.
| |
Collapse
|
32
|
Lunardi N, Oklopcic A, Prillaman M, Erisir A, Jevtovic-Todorovic V. Early Exposure to General Anesthesia Disrupts Spatial Organization of Presynaptic Vesicles in Nerve Terminals of the Developing Rat Subiculum. Mol Neurobiol 2015; 52:942-51. [PMID: 26048670 DOI: 10.1007/s12035-015-9246-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Indexed: 01/08/2023]
Abstract
Exposure to general anesthesia (GA) during critical stages of brain development induces widespread neuronal apoptosis and causes long-lasting behavioral deficits in numerous animal species. Although several studies have focused on the morphological fate of neurons dying acutely by GA-induced developmental neuroapoptosis, the effects of an early exposure to GA on the surviving synapses remain unclear. The aim of this study is to study whether exposure to GA disrupts the fine regulation of the dynamic spatial organization and trafficking of synaptic vesicles in presynaptic terminals. We exposed postnatal day 7 (PND7) rat pups to a clinically relevant anesthetic combination of midazolam, nitrous oxide, and isoflurane and performed a detailed ultrastructural analysis of the synaptic vesicle architecture at presynaptic terminals in the subiculum of rats at PND 12. In addition to a significant decrease in the density of presynaptic vesicles, we observed a reduction of docked vesicles, as well as a reduction of vesicles located within 100 nm from the active zone, in animals 5 days after an initial exposure to GA. We also found that the synaptic vesicles of animals exposed to GA are located more distally with respect to the plasma membrane than those of sham control animals and that the distance between presynaptic vesicles is increased in GA-exposed animals compared to sham controls. We report that exposure of immature rats to GA during critical stages of brain development causes significant disruption of the strategic topography of presynaptic vesicles within the nerve terminals of the subiculum.
Collapse
Affiliation(s)
- N Lunardi
- Department of Anesthesiology, University of Virginia Health System, PO Box 800710, Charlottesville, VA, 22908, USA,
| | | | | | | | | |
Collapse
|
33
|
Lin EP, Lee JR, Loepke AW. Anesthetics and the Developing Brain: The Yin and Yang. CURRENT ANESTHESIOLOGY REPORTS 2015. [DOI: 10.1007/s40140-015-0107-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
34
|
Ganella DE, Kim JH. Developmental rodent models of fear and anxiety: from neurobiology to pharmacology. Br J Pharmacol 2014; 171:4556-74. [PMID: 24527726 DOI: 10.1111/bph.12643] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 01/21/2014] [Accepted: 02/06/2014] [Indexed: 01/15/2023] Open
Abstract
Anxiety disorders pose one of the biggest threats to mental health in the world, and they predominantly emerge early in life. However, research of anxiety disorders and fear-related memories during development has been largely neglected, and existing treatments have been developed based on adult models of anxiety. The present review describes animal models of anxiety disorders across development and what is currently known of their pharmacology. To summarize, the underlying mechanisms of intrinsic 'unlearned' fear are poorly understood, especially beyond the period of infancy. Models using 'learned' fear reveal that through development, rats exhibit a stress hyporesponsive period before postnatal day 10, where they paradoxically form odour-shock preferences, and then switch to more adult-like conditioned fear responses. Juvenile rats appear to forget these aversive associations more easily, as is observed with the phenomenon of infantile amnesia. Juvenile rats also undergo more robust extinction, until adolescence where they display increased resistance to extinction. Maturation of brain structures, such as the amygdala, prefrontal cortex and hippocampus, along with the different temporal recruitment and involvement of various neurotransmitter systems (including NMDA, GABA, corticosterone and opioids) are responsible for these developmental changes. Taken together, the studies described in this review highlight that there is a period early in development where rats appear to be more robust in overcoming adverse early life experience. We need to understand the fundamental pharmacological processes underlying anxiety early in life in order to take advantage of this period for the treatment of anxiety disorders.
Collapse
Affiliation(s)
- Despina E Ganella
- Behavioural Neuroscience Division, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | | |
Collapse
|
35
|
Prenatal and postnatal development of synapses and acetylcholinesterase staining in the dentate gyrus of the rhesus monkey. Int J Dev Neurosci 2014; 1:77-97. [PMID: 24875720 DOI: 10.1016/0736-5748(83)90035-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/1982] [Indexed: 11/23/2022] Open
Abstract
Morphogenesis, distribution of cholinergic enzyme acetylcholinesterase and synaptogenesis in the dentate gyrus of the rhesus monkey during the pre- and postnatal periods of development were examined using histological, histochemical and ultrastructural methods. The pattern of neuronal differentiation in the dentate gyrus demonstrated distinct superficial-to-deep and lateral-to-medial gradients. The histochemical reaction for acetylcholinesterase was present on gestation day 120 as minimal staining in the supragranular band and in the inner one-third of the dentate molecular layer. At term, the laminar distribution of the enzyme assumed mature pattern although considerable enhancement in staining intensity was achieved postnatally. At term and at 9 months of postnatal age, the most pronounced enzyme activity was found in the supragranular band and in the inner one-third of the molecular layer. Synaptogenesis in the dentate molecular layer was characterized by the early formation of axo-dendritic contacts on dendritic trunks and branches followed by the appearance of synapses on simple and complex spines. Spines were detected infrequently on gestation day 132. On day 148, they ranged in morphology from short stubby protrusions to pedunculated, triangular processes. The majority of the spines exhibited flat postsynaptic surfaces. Complex, synapse-bearing U- and W-shaped spines were observed rarely at this age but appeared more frequently at term and at 15 months of postnatal age. However, at all ages, including 15 months postnatally, synapses on flat-surfaced simple spines predominated. Most synapses were of the asymmetric variety. With certain exceptions, these features of development of the rhesus dentate gyrus resemble the reported patterns of postnatal ontogenesis of this structure in the rat. However, the ingrowth of cholinergic afferents and the major modifications in synapse structure occur prenatally in the rhesus monkey during the second half of the gestation period. This temporal difference between the two species should receive consideration in the planning of neuroplasticity experiments designed to explore lesion-induced adaptations in afferent growth and synaptogenesis in the rhesus dentate gyrus.
Collapse
|
36
|
Bachevalier J, Beauregard M. Maturation of medial temporal lobe memory functions in rodents, monkeys, and humans. Hippocampus 2013. [DOI: 10.1002/hipo.1993.4500030723] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Jocelyne Bachevalier
- University of Texas Medical School, Department of Neurobiology and Anatomy, Houston, Texas, U.S.A
| | - Mario Beauregard
- University of Texas Medical School, Department of Neurobiology and Anatomy, Houston, Texas, U.S.A
| |
Collapse
|
37
|
Semple BD, Blomgren K, Gimlin K, Ferriero DM, Noble-Haeusslein LJ. Brain development in rodents and humans: Identifying benchmarks of maturation and vulnerability to injury across species. Prog Neurobiol 2013; 106-107:1-16. [PMID: 23583307 PMCID: PMC3737272 DOI: 10.1016/j.pneurobio.2013.04.001] [Citation(s) in RCA: 1496] [Impact Index Per Article: 124.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 04/03/2013] [Accepted: 04/03/2013] [Indexed: 12/13/2022]
Abstract
Hypoxic-ischemic and traumatic brain injuries are leading causes of long-term mortality and disability in infants and children. Although several preclinical models using rodents of different ages have been developed, species differences in the timing of key brain maturation events can render comparisons of vulnerability and regenerative capacities difficult to interpret. Traditional models of developmental brain injury have utilized rodents at postnatal day 7-10 as being roughly equivalent to a term human infant, based historically on the measurement of post-mortem brain weights during the 1970s. Here we will examine fundamental brain development processes that occur in both rodents and humans, to delineate a comparable time course of postnatal brain development across species. We consider the timing of neurogenesis, synaptogenesis, gliogenesis, oligodendrocyte maturation and age-dependent behaviors that coincide with developmentally regulated molecular and biochemical changes. In general, while the time scale is considerably different, the sequence of key events in brain maturation is largely consistent between humans and rodents. Further, there are distinct parallels in regional vulnerability as well as functional consequences in response to brain injuries. With a focus on developmental hypoxic-ischemic encephalopathy and traumatic brain injury, this review offers guidelines for researchers when considering the most appropriate rodent age for the developmental stage or process of interest to approximate human brain development.
Collapse
Affiliation(s)
- Bridgette D. Semple
- Department of Neurological Surgery, University of California San Francisco, 513 Parnassus Avenue, Room HSE-722, San Francisco, CA 94143-0112, USA
| | - Klas Blomgren
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Sweden
- Department of Pediatrics, Queen Silvia's Children's Hospital, University of Gothenburg, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Q2:07, SE 171 76 Stockholm, Sweden
| | - Kayleen Gimlin
- Department of Neurological Surgery, University of California San Francisco, 513 Parnassus Avenue, Room HSE-722, San Francisco, CA 94143-0112, USA
| | - Donna M. Ferriero
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Linda J. Noble-Haeusslein
- Department of Neurological Surgery, University of California San Francisco, 513 Parnassus Avenue, Room HSE-722, San Francisco, CA 94143-0112, USA
- Department of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
38
|
Snyder MA, Adelman AE, Gao WJ. Gestational methylazoxymethanol exposure leads to NMDAR dysfunction in hippocampus during early development and lasting deficits in learning. Neuropsychopharmacology 2013; 38:328-40. [PMID: 22968815 PMCID: PMC3527110 DOI: 10.1038/npp.2012.180] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The N-methyl-D-aspartate (NMDA) receptor has long been associated with learning and memory processes as well as diseased states, particularly in schizophrenia (SZ). Additionally, SZ is increasingly recognized as a neurodevelopmental disorder with cognitive impairments often preceding the onset of psychosis. However, the cause of these cognitive deficits and what initiates the pathological process is unknown. Growing evidence has implicated the glutamate system and, in particular, N-methyl-D-aspartate receptor (NMDAR) dysfunction in the pathophysiology of SZ. Yet, the vast majority of SZ-related research has focused on NMDAR function in adults leaving the role of NMDARs during development uncharacterized. We used the prenatal methylazoxymethanol acetate (MAM, E17) exposure model to determine the alterations of NMDAR protein levels and function, as well as associated cognitive deficits during development. We found that MAM-exposed animals have significantly altered NMDAR protein levels and function in the juvenile and adolescent hippocampus. Furthermore, these changes are associated with learning and memory deficits in the Morris Water Maze. Thus, in the prenatal MAM-exposure SZ model, NMDAR expression and function is altered during the critical period of hippocampal development. These changes may be involved in disease initiation and cognitive impairment in the early stage of SZ.
Collapse
Affiliation(s)
- Melissa A Snyder
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Alicia E Adelman
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA,Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA, Tel: +215 991 8907, Fax: +215 843 9802, E-mail:
| |
Collapse
|
39
|
Helfer JL, White ER, Christie BR. Enhanced deficits in long-term potentiation in the adult dentate gyrus with 2nd trimester ethanol consumption. PLoS One 2012; 7:e51344. [PMID: 23227262 PMCID: PMC3515437 DOI: 10.1371/journal.pone.0051344] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 11/05/2012] [Indexed: 12/05/2022] Open
Abstract
Ethanol exposure during pregnancy can cause structural and functional changes in the brain that can impair cognitive capacity. The hippocampal formation, an area of the brain strongly linked with learning and memory, is particularly vulnerable to the teratogenic effects of ethanol. In the present experiments we sought to determine if the functional effects of developmental ethanol exposure could be linked to ethanol exposure during any single trimester-equivalent. Ethanol exposure during the 1st or 3rd trimester-equivalent produced only minor changes in synaptic plasticity in adult offspring. In contrast, ethanol exposure during the 2nd trimester equivalent resulted in a pronounced decrease in long-term potentiation, indicating that the timing of exposure influences the severity of the deficit. Together, the results from these experiments demonstrate long-lasting alterations in synaptic plasticity as the result of developmental ethanol exposure and dependent on the timing of exposure. Furthermore, these results allude to neural circuit malfunction within the hippocampal formation, perhaps relating to the learning and memory deficits observed in individuals with fetal alcohol spectrum disorders.
Collapse
Affiliation(s)
- Jennifer L. Helfer
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Department of Biology, University of Victoria, Victoria, British Columbia, Canada
| | - Emily R. White
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Graduate Program in Neuroscience, University of Victoria, Victoria, British Columbia, Canada
| | - Brian R. Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Department of Biology, University of Victoria, Victoria, British Columbia, Canada
- Graduate Program in Neuroscience, University of Victoria, Victoria, British Columbia, Canada
- Graduate Program in Neuroscience and the Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- The Brain Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
40
|
Abstract
Bioluminescence imaging (BLI) takes advantage of the light-emitting properties of luciferase enzymes, which produce light upon oxidizing a substrate (i.e., D-luciferin) in the presence of molecular oxygen and energy. Photons emitted from living tissues can be detected and quantified by a highly sensitive charge-coupled device camera, enabling the investigator to noninvasively analyze the dynamics of biomolecular reactions in a variety of living model organisms such as transgenic mice. BLI has been used extensively in cancer research, cell transplantation, and for monitoring of infectious diseases, but only recently experimental models have been designed to study processes and pathways in neurological disorders such as Alzheimer disease, Parkinson disease, or amyotrophic lateral sclerosis. In this review, we highlight recent applications of BLI in neuroscience, including transgene expression in the brain, longitudinal studies of neuroinflammatory responses to neurodegeneration and injury, and in vivo imaging studies of neurogenesis and mitochondrial toxicity. Finally, we highlight some new developments of BLI compounds and luciferase substrates with promising potential for in vivo studies of neurological dysfunctions.
Collapse
Affiliation(s)
- Katja Hochgräfe
- DZNE (German Center for Neurodegenerative Diseases), Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
| | | |
Collapse
|
41
|
Armstrong BC, Le Boutillier JC, Petit TL. Ultrastructural synaptic changes associated with neurofibromatosis type 1: a quantitative analysis of hippocampal region CA1 in a Nf1(+/-) mouse model. Synapse 2011; 66:246-55. [PMID: 22121000 DOI: 10.1002/syn.21507] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 10/28/2011] [Accepted: 11/02/2011] [Indexed: 11/06/2022]
Abstract
Neurofibromatosis type 1 (NF1) is one of the most frequently diagnosed autosomal dominant inherited disorders resulting in neurological dysfunction, including an assortment of learning disabilities and cognitive deficits. To elucidate the neural mechanisms underlying the disorder, we employed a mouse model (Nf1(+/-) ) to conduct a quantitative analysis of ultrastructural changes associated with the NF1 disorder. Using both serial light and electron microscopy, we examined reconstructions of the CA1 region of the hippocampus, which is known to play a central role in many of the dysfunctions associated with NF1. In general, the morphology of synapses in both the Nf1(+/-) and wild-type groups of animals were similar. No differences were observed in synapse per neuron density, pre- and postsynaptic areas, or lengths. However, concave synapses were found to show a lower degree of curvature in the Nf1(+/-) mutant than in the wild type. These results indicate that the synaptic ultrastructure of Nf1(+/-) mice appears relatively normal with the exception of the degree of synaptic curvature in concave synapses, adding further support to the importance of synaptic curvature in synaptic plasticity, learning, and memory.
Collapse
Affiliation(s)
- Blair C Armstrong
- Department of Psychology and Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, USA
| | | | | |
Collapse
|
42
|
Sanchez V, Feinstein SD, Lunardi N, Joksovic PM, Boscolo A, Todorovic SM, Jevtovic-Todorovic V. General Anesthesia Causes Long-term Impairment of Mitochondrial Morphogenesis and Synaptic Transmission in Developing Rat Brain. Anesthesiology 2011; 115:992-1002. [PMID: 21909020 PMCID: PMC3203321 DOI: 10.1097/aln.0b013e3182303a63] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Clinically used general anesthetics, alone or in combination, are damaging to the developing mammalian brain. In addition to causing widespread apoptotic neurodegeneration in vulnerable brain regions, exposure to general anesthesia at the peak of synaptogenesis causes learning and memory deficiencies later in life. In vivo rodent studies have suggested that activation of the intrinsic (mitochondria-dependent) apoptotic pathway is the earliest warning sign of neuronal damage, suggesting that a disturbance in mitochondrial integrity and function could be the earliest triggering events. METHODS Because proper and timely mitochondrial morphogenesis is critical for brain development, the authors examined the long-term effects of a commonly used anesthesia combination (isoflurane, nitrous oxide, and midazolam) on the regional distribution, ultrastructural properties, and electron transport chain function of mitochondria, as well as synaptic neurotransmission, in the subiculum of rat pups. RESULTS This anesthesia, administered at the peak of synaptogenesis, causes protracted injury to mitochondria, including significant enlargement of mitochondria (more than 30%, P < 0.05), impairment of their structural integrity, an approximately 28% increase in their complex IV activity (P < 0.05), and a twofold decrease in their regional distribution in presynaptic neuronal profiles (P < 0.05), where their presence is important for the normal development and functioning of synapses. Consequently, the authors showed that impaired mitochondrial morphogenesis is accompanied by heightened autophagic activity, decrease in mitochondrial density (approximately 27%, P < 0.05), and long-lasting disturbances in inhibitory synaptic neurotransmission. The interrelation of these phenomena remains to be established. CONCLUSION Developing mitochondria are exquisitely vulnerable to general anesthesia and may be important early target of anesthesia-induced developmental neurodegeneration.
Collapse
Affiliation(s)
- Victoria Sanchez
- Dept. of Anesthesiology, University of Virginia, Charlottesville, Virginia
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia
| | - Shawn D. Feinstein
- Dept. of Anesthesiology, University of Virginia, Charlottesville, Virginia
| | - Nadia Lunardi
- Dept. of Anesthesiology, University of Virginia, Charlottesville, Virginia
- Dept. of Anesthesiology and Pharmacology, University of Padova, Padova, Italy
| | - Pavle M. Joksovic
- Dept. of Anesthesiology, University of Virginia, Charlottesville, Virginia
- Dept. of Psychiatry, Yale University, New Haven, Connecticut
| | - Annalisa Boscolo
- Dept. of Anesthesiology, University of Virginia, Charlottesville, Virginia
- Dept. of Anesthesiology and Pharmacology, University of Padova, Padova, Italy
| | - Slobodan M. Todorovic
- Dept. of Anesthesiology, University of Virginia, Charlottesville, Virginia
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia
| | - Vesna Jevtovic-Todorovic
- Dept. of Anesthesiology, University of Virginia, Charlottesville, Virginia
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
43
|
Abstract
Anesthesia kills neurons in the brain of infantile animals, including primates, and causes permanent and progressive neurocognitive decline. The anesthesia community and regulatory authorities alike are concerned that is also true in humans. In this review, I summarize what we currently know about the risks of pediatric anesthesia to long-term cognitive function. If anesthesia is discovered to cause cognitive decline in humans, we need to know how to prevent and treat it. Prevention requires knowledge of the mechanisms of anesthesia-induced cognitive decline. This review gives an overview of some of the mechanisms that have been proposed for anesthesia-induced cognitive decline and discusses possible treatment options. If anesthesia induces cognitive decline in humans, we need to know what type and duration of anesthetic is safe, and which, if any, is not safe. This review discusses early results of comparative animal studies of anesthetic neurotoxicity. Until we know if and how pediatric anesthesia affects cognition in humans, a change in anesthetic practice would be premature, not guided by evidence of better alternatives, and therefore potentially dangerous. The SmartTots initiative jointly supported by the International Anesthesia Research Society and the Food and Drug Administration aims to fund research designed to shed light on these issues that are of high priority to the anesthesia community and the public alike and therefore deserves the full support of these interest groups.
Collapse
Affiliation(s)
- Greg Stratmann
- Department of Anesthesia and Perioperative Care, University of California San Francisco, Box 0464, Room U286, 513 Parnassus Ave., San Francisco, CA 94143, USA.
| |
Collapse
|
44
|
Diamantopoulou A, Stamatakis A, Panagiotaropoulos T, Stylianopoulou F. Reward or its denial during the neonatal period affects adult spatial memory and hippocampal phosphorylated cAMP response element-binding protein levels of both the neonatal and adult rat. Neuroscience 2011; 181:89-99. [DOI: 10.1016/j.neuroscience.2011.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 03/01/2011] [Accepted: 03/01/2011] [Indexed: 01/03/2023]
|
45
|
|
46
|
Dendritic spines and development: towards a unifying model of spinogenesis--a present day review of Cajal's histological slides and drawings. Neural Plast 2011; 2010:769207. [PMID: 21584262 PMCID: PMC3091278 DOI: 10.1155/2010/769207] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Accepted: 11/14/2010] [Indexed: 12/21/2022] Open
Abstract
Dendritic spines receive the majority of excitatory connections in the central nervous system, and, thus, they are key structures in the regulation of neural activity. Hence, the cellular and molecular mechanisms underlying their generation and plasticity, both during development and in adulthood, are a matter of fundamental and practical interest. Indeed, a better understanding of these mechanisms should provide clues to the development of novel clinical therapies. Here, we present original results obtained from high-quality images of Cajal's histological preparations, stored at the Cajal Museum (Instituto Cajal, CSIC), obtained using extended focus imaging, three-dimensional reconstruction, and rendering. Based on the data available in the literature regarding the formation of dendritic spines during development and our results, we propose a unifying model for dendritic spine development.
Collapse
|
47
|
Grossman AW, Aldridge GM, Lee KJ, Zeman MK, Jun CS, Azam HS, Arii T, Imoto K, Greenough WT, Rhyu IJ. Developmental characteristics of dendritic spines in the dentate gyrus of Fmr1 knockout mice. Brain Res 2010; 1355:221-7. [PMID: 20682298 PMCID: PMC3433497 DOI: 10.1016/j.brainres.2010.07.090] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2010] [Revised: 07/24/2010] [Accepted: 07/27/2010] [Indexed: 11/23/2022]
Abstract
Fragile X Syndrome (FXS) is the most common form of inherited mental retardation. The neuroanatomical phenotype of adult FXS patients, as well as adult Fmr1 knockout (KO) mice, includes elevated dendritic spine density and a spine morphology profile in neocortex that resembles younger individuals. Developmental studies in mouse neocortex have revealed a dynamic phenotype that varies with age, especially during the period of synaptic pruning. Here we investigated the hippocampal dentate gyrus to determine if the FXS spine phenotype is similarly tied to periods of maturation and pruning in this brain region. We used high-voltage electron microscopy to characterize Golgi-stained spines along granule cell dendrites in Fmr1 KO and wildtype (WT) mouse dentate gyrus at postnatal days 15, 21, 30, and 60. In contrast to neocortex, dendritic spine density was higher in Fmr1 KO mice across development. Interestingly, neither genotype showed specific phases of synaptogenesis or pruning, potentially explaining the phenotypic differences from neocortex. Similarly, although the KO mice showed a more immature morphological phenotype overall than WT (higher proportion of thin headed spines, lower proportion of mushroom and stubby spines), both genotypes showed gradual development, rather than impairments during specific phases of maturation. Finally, spine length showed a complex developmental pattern that differs from other brain regions examined, suggesting dynamic regulation by FMRP and other brain region-specific proteins. These findings shed new light on FMRP's role in development and highlight the need for new techniques to further understand the mechanisms by which FMRP affects synaptic maturation.
Collapse
Affiliation(s)
- Aaron W. Grossman
- Beckman Institute, University of Illinois at Urbana-Champaign, USA
- Neuroscience Graduate Program, University of Illinois at Urbana-Champaign, USA
- Medical Scholars Program, University of Illinois at Urbana-Champaign, USA
| | - Georgina M. Aldridge
- Beckman Institute, University of Illinois at Urbana-Champaign, USA
- Neuroscience Graduate Program, University of Illinois at Urbana-Champaign, USA
- Medical Scholars Program, University of Illinois at Urbana-Champaign, USA
| | - Kea Joo Lee
- Section of Brain Structure Information, Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan
| | | | - Christine S. Jun
- Beckman Institute, University of Illinois at Urbana-Champaign, USA
| | - Humza S. Azam
- Beckman Institute, University of Illinois at Urbana-Champaign, USA
| | - Tatsuo Arii
- Section of Brain Structure Information, Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan
| | - Keiji Imoto
- Department of Information Physiology, National Institute for Physiological Sciences, Okazaki 444-8787, Japan
| | - William T. Greenough
- Beckman Institute, University of Illinois at Urbana-Champaign, USA
- Neuroscience Graduate Program, University of Illinois at Urbana-Champaign, USA
- Medical Scholars Program, University of Illinois at Urbana-Champaign, USA
- Departments of Psychology, Psychiatry, and Cell and Structural Biology, University of Illinois at Urbana-Champaign, USA
| | - Im Joo Rhyu
- Beckman Institute, University of Illinois at Urbana-Champaign, USA
- Department of Anatomy, Korea University College of Medicine, Seoul, 136-705, Korea
| |
Collapse
|
48
|
Raineki C, Holman PJ, Debiec J, Bugg M, Beasley A, Sullivan RM. Functional emergence of the hippocampus in context fear learning in infant rats. Hippocampus 2010; 20:1037-46. [PMID: 19739248 PMCID: PMC2891848 DOI: 10.1002/hipo.20702] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The hippocampus is a part of the limbic system and is important for the formation of associative memories, such as acquiring information about the context (e.g., the place where an experience occurred) during emotional learning (e.g., fear conditioning). Here, we assess whether the hippocampus is responsible for pups' newly emerging context learning. In all experiments, postnatal day (PN) 21 and PN24 rat pups received 10 pairings of odor-0.5 mA shock or control unpaired odor-shock, odor only, or shock only. Some pups were used for context, cue or odor avoidance tests, while the remaining pups were used for c-Fos immunohistochemistry to assess hippocampal activity during acquisition. Our results show that cue and odor avoidance learning were similar at both ages, while contextual fear learning and learning-associated hippocampal (CA1, CA3, and dentate gyrus) activity (c-Fos) only occurred in PN24 paired pups. To assess a causal relationship between the hippocampus and context conditioning, we infused muscimol into the hippocampus, which blocked acquisition of context fear learning in the PN24 pups. Muscimol or vehicle infusions did not affect cue learning or aversion to the odor at PN21 or PN24. The results suggest that the newly emerging contextual learning exhibited by PN24 pups is supported by the hippocampus.
Collapse
Affiliation(s)
- Charlis Raineki
- Emotional Brain Institute, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Swayne LA, Sorbara CD, Bennett SAL. Pannexin 2 is expressed by postnatal hippocampal neural progenitors and modulates neuronal commitment. J Biol Chem 2010; 285:24977-86. [PMID: 20529862 DOI: 10.1074/jbc.m110.130054] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The pannexins (Panx1, -2, and -3) are a mammalian family of putative single membrane channels discovered through homology to invertebrate gap junction-forming proteins, the innexins. Because connexin gap junction proteins are known regulators of neural stem and progenitor cell proliferation, migration, and specification, we asked whether pannexins, specifically Panx2, play a similar role in the postnatal hippocampus. We show that Panx2 protein is differentially expressed by multipotential progenitor cells and mature neurons. Both in vivo and in vitro, Type I and IIa stem-like neural progenitor cells express an S-palmitoylated Panx2 species localizing to Golgi and endoplasmic reticulum membranes. Protein expression is down-regulated during neurogenesis in neuronally committed Type IIb and III progenitor cells and immature neurons. Panx2 is re-expressed by neurons following maturation. Protein expressed by mature neurons is not palmitoylated and localizes to the plasma membrane. To assess the impact of Panx2 on neuronal differentiation, we used short hairpin RNA to suppress Panx2 expression in Neuro2a cells. Knockdown significantly accelerated the rate of neuronal differentiation. Neuritic extension and the expression of antigenic markers of mature neurons occurred earlier in stable lines expressing Panx2 short hairpin RNA than in controls. Together, these findings describe an endogenous post-translational regulation of Panx2, specific to early neural progenitor cells, and demonstrate that this expression plays a role in modulating the timing of their commitment to a neuronal lineage.
Collapse
Affiliation(s)
- Leigh Anne Swayne
- Department of Biochemistry, Neural Regeneration Laboratory and Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1M 1E5, Canada.
| | | | | |
Collapse
|
50
|
Thind KK, Yamawaki R, Phanwar I, Zhang G, Wen X, Buckmaster PS. Initial loss but later excess of GABAergic synapses with dentate granule cells in a rat model of temporal lobe epilepsy. J Comp Neurol 2010; 518:647-67. [PMID: 20034063 DOI: 10.1002/cne.22235] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Many patients with temporal lobe epilepsy display neuron loss in the dentate gyrus. One potential epileptogenic mechanism is loss of GABAergic interneurons and inhibitory synapses with granule cells. Stereological techniques were used to estimate numbers of gephyrin-positive punctae in the dentate gyrus, which were reduced short-term (5 days after pilocarpine-induced status epilepticus) but later rebounded beyond controls in epileptic rats. Stereological techniques were used to estimate numbers of synapses in electron micrographs of serial sections processed for postembedding GABA-immunoreactivity. Adjacent sections were used to estimate numbers of granule cells and glutamic acid decarboxylase-positive neurons per dentate gyrus. GABAergic neurons were reduced to 70% of control levels short-term, where they remained in epileptic rats. Integrating synapse and cell counts yielded average numbers of GABAergic synapses per granule cell, which decreased short-term and rebounded in epileptic animals beyond control levels. Axo-shaft and axo-spinous GABAergic synapse numbers in the outer molecular layer changed most. These findings suggest interneuron loss initially reduces numbers of GABAergic synapses with granule cells, but later, synaptogenesis by surviving interneurons overshoots control levels. In contrast, the average number of excitatory synapses per granule cell decreased short-term but recovered only toward control levels, although in epileptic rats excitatory synapses in the inner molecular layer were larger than in controls. These findings reveal a relative excess of GABAergic synapses and suggest that reports of reduced functional inhibitory synaptic input to granule cells in epilepsy might be attributable not to fewer but instead to abundant but dysfunctional GABAergic synapses.
Collapse
Affiliation(s)
- Khushdev K Thind
- Department of Comparative Medicine, Stanford University, California 94305, USA
| | | | | | | | | | | |
Collapse
|