1
|
Svirsky SE, Henchir J, Li Y, Carlson SW, Dixon CE. Temporal-Specific Sex and Injury-Dependent Changes on Neurogranin-Associated Synaptic Signaling After Controlled Cortical Impact in Rats. Mol Neurobiol 2024; 61:7256-7268. [PMID: 38376763 DOI: 10.1007/s12035-024-04043-5] [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: 09/08/2023] [Accepted: 02/08/2024] [Indexed: 02/21/2024]
Abstract
Extensive effort has been made to study the role of synaptic deficits in cognitive impairment after traumatic brain injury (TBI). Neurogranin (Ng) is a calcium-sensitive calmodulin (CaM)-binding protein essential for Ca2+/CaM-dependent kinase II (CaMKII) autophosphorylation which subsequently modulates synaptic plasticity. Given the loss of Ng expression after injury, additional research is warranted to discern changes in hippocampal post-synaptic signaling after TBI. Under isoflurane anesthesia, adult, male and female Sprague-Dawley rats received a sham/control or controlled cortical impact (CCI) injury. Ipsilateral hippocampal synaptosomes were isolated at 24 h and 1, 2, and 4 weeks post-injury, and western blot was used to evaluate protein expression of Ng-associated signaling proteins. Non-parametric Mann-Whitney tests were used to determine significance of injury for each sex at each time point. There were significant changes in the hippocampal synaptic expression of Ng and associated synaptic proteins such as phosphorylated Ng, CaMKII, and CaM up to 4 weeks post-CCI, demonstrating TBI alters hippocampal post-synaptic signaling. This study furthers our understanding of mechanisms of cognitive dysfunction within the synapse sub-acutely after TBI.
Collapse
Affiliation(s)
- Sarah E Svirsky
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Jeremy Henchir
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Youming Li
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Shaun W Carlson
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - C Edward Dixon
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA.
- V.A. Pittsburgh Healthcare System, Pittsburgh, PA, USA.
| |
Collapse
|
2
|
Alkadhi KA. Synaptic Plasticity and Cognitive Ability in Experimental Adult-Onset Hypothyroidism. J Pharmacol Exp Ther 2024; 389:150-162. [PMID: 38508752 DOI: 10.1124/jpet.123.001887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 02/05/2024] [Accepted: 02/20/2024] [Indexed: 03/22/2024] Open
Abstract
Adult-onset hypothyroidism impairs normal brain function. Research on animal models of hypothyroidism has revealed critical information on how deficiency of thyroid hormones impacts the electrophysiological and molecular functions of the brain, which leads to the well known cognitive impairment in untreated hypothyroid patients. Currently, such information can only be obtained from experiments on animal models of hypothyroidism. This review summarizes important research findings that pertain to understanding the clinical cognitive consequences of hypothyroidism, which will provide a better guiding path for therapy of hypothyroidism. SIGNIFICANCE STATEMENT: Cognitive impairment occurs during adult-onset hypothyroidism in both humans and animal models. Findings from animal studies validate clinical findings showing impaired long-term potentiation, decreased CaMKII, and increased calcineurin. Such findings can only be gleaned from animal experiments to show how hypothyroidism produces clinical symptoms.
Collapse
Affiliation(s)
- Karim A Alkadhi
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| |
Collapse
|
3
|
Alba-González A, Yáñez J, Anadón R, Folgueira M. Neurogranin-like immunoreactivity in the zebrafish brain during development. Brain Struct Funct 2022; 227:2593-2607. [PMID: 36018391 PMCID: PMC9618489 DOI: 10.1007/s00429-022-02550-6] [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: 05/10/2022] [Accepted: 08/03/2022] [Indexed: 11/30/2022]
Abstract
Neurogranin (Nrgn) is a neural protein that is enriched in the cerebral cortex and is involved in synaptic plasticity via its interaction with calmodulin. Recently we reported its expression in the brain of the adult zebrafish (Alba-González et al. J Comp Neurol 530:1569–1587, 2022). In this study we analyze the development of Nrgn-like immunoreactivity (Nrgn-like-ir) in the brain and sensory structures of zebrafish embryos and larvae, using whole mounts and sections. First Nrgn-like positive neurons appeared by 2 day post-fertilization (dpf) in restricted areas of the brain, mostly in the pallium, epiphysis and hindbrain. Nrgn-like populations increased noticeably by 3 dpf, reaching an adult-like pattern in 6 dpf. Most Nrgn-like positive neurons were observed in the olfactory organ, retina (most ganglion cells, some amacrine and bipolar cells), pallium, lateral hypothalamus, thalamus, optic tectum, torus semicircularis, octavolateralis area, and viscerosensory column. Immunoreactivity was also observed in axonal tracts originating in Nrgn-like neuronal populations, namely, the projection of Nrgn-like immunopositive primary olfactory fibers to olfactory glomeruli, that of Nrgn-like positive pallial cells to the hypothalamus, the Nrgn-like-ir optic nerve to the pretectum and optic tectum, the Nrgn-like immunolabeled lateral hypothalamus to the contralateral region via the horizontal commissure, the octavolateralis area to the midbrain via the lateral lemniscus, and the viscerosensory column to the dorsal isthmus via the secondary gustatory tract. The late expression of Nrgn in zebrafish neurons is probably related to functional maturation of higher brain centers, as reported in the mammalian telencephalon. The analysis of Nrgn expression in the zebrafish brain suggests that it may be a useful marker for specific neuronal circuitries.
Collapse
Affiliation(s)
- Anabel Alba-González
- Department of Biology, Faculty of Sciences, University of A Coruña, Campus da Zapateira, 15008-A, Coruña, Spain.,Centro de Investigaciones Científicas Avanzadas (CICA), University of A Coruña, 15071-A, Coruña, Spain
| | - Julián Yáñez
- Department of Biology, Faculty of Sciences, University of A Coruña, Campus da Zapateira, 15008-A, Coruña, Spain. .,Centro de Investigaciones Científicas Avanzadas (CICA), University of A Coruña, 15071-A, Coruña, Spain.
| | - Ramón Anadón
- Department of Functional Biology, Faculty of Biology, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Mónica Folgueira
- Department of Biology, Faculty of Sciences, University of A Coruña, Campus da Zapateira, 15008-A, Coruña, Spain. .,Centro de Investigaciones Científicas Avanzadas (CICA), University of A Coruña, 15071-A, Coruña, Spain.
| |
Collapse
|
4
|
Svirsky S, Henchir J, Li Y, Ma X, Carlson S, Dixon CE. Neurogranin Protein Expression Is Reduced after Controlled Cortical Impact in Rats. J Neurotrauma 2020; 37:939-949. [PMID: 31691647 PMCID: PMC7175627 DOI: 10.1089/neu.2019.6759] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Traumatic brain injury (TBI) is known to cause short- and long-term synaptic changes in the brain, possibly underlying downstream cognitive impairments. Neuronal levels of neurogranin, a calcium-sensitive calmodulin-binding protein essential for synaptic plasticity and postsynaptic signaling, are correlated with cognitive function. This study aims to understand the effect of TBI on neurogranin by characterizing changes in protein expression at various time points after injury. Adult, male rats were subjected to either controlled cortical impact (CCI) or control surgery. Expression of neurogranin and post-synaptic density 95 (PSD-95) were evaluated by Western blot in the cortex and hippocampus at 24 h and 1, 2, and 4 weeks post-injury. We hypothesized that CCI reduces neurogranin levels in the cortex and hippocampus, and demonstrate different expression patterns from PSD-95. Neurogranin levels were reduced in the ipsilateral cortex and hippocampus up to 2 weeks after injury but recovered to sham levels by 4 weeks. The contralateral cortex and hippocampus were relatively resistant to changes in neurogranin expression post-injury. Qualitative immunohistochemical assessment corroborated the immunoblot findings. Particularly, the pericontusional cortex and ipsilateral Cornu Ammonis (CA)3 region showed marked reduction in immunoreactivity. PSD-95 demonstrated similar expression patterns to neurogranin in the cortex; however, in the hippocampus, protein expression was increased compared with sham at the 2 and 4 week time points. Our results indicate that CCI lowers neurogranin expression with temporal and regional specificity and that this occurs independently of dendritic loss. Further understanding of the role of neurogranin in synaptic biology after TBI will elucidate pathological mechanisms contributing to cognitive dysfunction.
Collapse
Affiliation(s)
- Sarah Svirsky
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Jeremy Henchir
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Youming Li
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Xiecheng Ma
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Shaun Carlson
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - C. Edward Dixon
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- V.A. Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| |
Collapse
|
5
|
Lewczuk P, Riederer P, O’Bryant SE, Verbeek MM, Dubois B, Visser PJ, Jellinger KA, Engelborghs S, Ramirez A, Parnetti L, Jack CR, Teunissen CE, Hampel H, Lleó A, Jessen F, Glodzik L, de Leon MJ, Fagan AM, Molinuevo JL, Jansen WJ, Winblad B, Shaw LM, Andreasson U, Otto M, Mollenhauer B, Wiltfang J, Turner MR, Zerr I, Handels R, Thompson AG, Johansson G, Ermann N, Trojanowski JQ, Karaca I, Wagner H, Oeckl P, van Waalwijk van Doorn L, Bjerke M, Kapogiannis D, Kuiperij HB, Farotti L, Li Y, Gordon BA, Epelbaum S, Vos SJB, Klijn CJM, Van Nostrand WE, Minguillon C, Schmitz M, Gallo C, Mato AL, Thibaut F, Lista S, Alcolea D, Zetterberg H, Blennow K, Kornhuber J, Riederer P, Gallo C, Kapogiannis D, Mato AL, Thibaut F. Cerebrospinal fluid and blood biomarkers for neurodegenerative dementias: An update of the Consensus of the Task Force on Biological Markers in Psychiatry of the World Federation of Societies of Biological Psychiatry. World J Biol Psychiatry 2018; 19:244-328. [PMID: 29076399 PMCID: PMC5916324 DOI: 10.1080/15622975.2017.1375556] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the 12 years since the publication of the first Consensus Paper of the WFSBP on biomarkers of neurodegenerative dementias, enormous advancement has taken place in the field, and the Task Force takes now the opportunity to extend and update the original paper. New concepts of Alzheimer's disease (AD) and the conceptual interactions between AD and dementia due to AD were developed, resulting in two sets for diagnostic/research criteria. Procedures for pre-analytical sample handling, biobanking, analyses and post-analytical interpretation of the results were intensively studied and optimised. A global quality control project was introduced to evaluate and monitor the inter-centre variability in measurements with the goal of harmonisation of results. Contexts of use and how to approach candidate biomarkers in biological specimens other than cerebrospinal fluid (CSF), e.g. blood, were precisely defined. Important development was achieved in neuroimaging techniques, including studies comparing amyloid-β positron emission tomography results to fluid-based modalities. Similarly, development in research laboratory technologies, such as ultra-sensitive methods, raises our hopes to further improve analytical and diagnostic accuracy of classic and novel candidate biomarkers. Synergistically, advancement in clinical trials of anti-dementia therapies energises and motivates the efforts to find and optimise the most reliable early diagnostic modalities. Finally, the first studies were published addressing the potential of cost-effectiveness of the biomarkers-based diagnosis of neurodegenerative disorders.
Collapse
Affiliation(s)
- Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, and Department of Biochemical Diagnostics, University Hospital of Białystok, Białystok, Poland
| | - Peter Riederer
- Center of Mental Health, Clinic and Policlinic of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Würzburg, Würzburg, Germany
| | - Sid E. O’Bryant
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Marcel M. Verbeek
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Center, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer center, Nijmegen, The Netherlands
| | - Bruno Dubois
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Salpêtrièrie Hospital, INSERM UMR-S 975 (ICM), Paris 6 University, Paris, France
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
- Department of Neurology, Alzheimer Centre, Amsterdam Neuroscience VU University Medical Centre, Amsterdam, The Netherlands
| | | | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), University of Antwerp, Antwerp, Belgium
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Alfredo Ramirez
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | - Lucilla Parnetti
- Section of Neurology, Center for Memory Disturbances, Lab of Clinical Neurochemistry, University of Perugia, Perugia, Italy
| | | | - Charlotte E. Teunissen
- Neurochemistry Lab and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Harald Hampel
- AXA Research Fund & UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
| | - Alberto Lleó
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Spain
| | - Frank Jessen
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
- German Center for Neurodegenerative Disorders (DZNE), Bonn, Germany
| | - Lidia Glodzik
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA
| | - Mony J. de Leon
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA
| | - Anne M. Fagan
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - José Luis Molinuevo
- Barcelonabeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
- Alzheimer’s Disease and Other Cognitive Disorders Unit, Hospital Clínic, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Willemijn J. Jansen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
| | - Bengt Winblad
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ulf Andreasson
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kassel and University Medical Center Göttingen, Department of Neurology, Göttingen, Germany
| | - Jens Wiltfang
- Department of Psychiatry & Psychotherapy, University of Göttingen, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- iBiMED, Medical Sciences Department, University of Aveiro, Aveiro, Portugal
| | - Martin R. Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Inga Zerr
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Clinical Dementia Centre, Department of Neurology, University Medical School, Göttingen, Germany
| | - Ron Handels
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden
| | | | - Gunilla Johansson
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden
| | - Natalia Ermann
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - John Q. Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilker Karaca
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Holger Wagner
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Patrick Oeckl
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Linda van Waalwijk van Doorn
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Center, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer center, Nijmegen, The Netherlands
| | - Maria Bjerke
- Reference Center for Biological Markers of Dementia (BIODEM), University of Antwerp, Antwerp, Belgium
| | - Dimitrios Kapogiannis
- Laboratory of Neurosciences, National Institute on Aging/National Institutes of Health (NIA/NIH), Baltimore, MD, USA
| | - H. Bea Kuiperij
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Center, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer center, Nijmegen, The Netherlands
| | - Lucia Farotti
- Section of Neurology, Center for Memory Disturbances, Lab of Clinical Neurochemistry, University of Perugia, Perugia, Italy
| | - Yi Li
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA
| | - Brian A. Gordon
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Stéphane Epelbaum
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Salpêtrièrie Hospital, INSERM UMR-S 975 (ICM), Paris 6 University, Paris, France
| | - Stephanie J. B. Vos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
| | - Catharina J. M. Klijn
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Center, Nijmegen, The Netherlands
| | | | - Carolina Minguillon
- Barcelonabeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Matthias Schmitz
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Clinical Dementia Centre, Department of Neurology, University Medical School, Göttingen, Germany
| | - Carla Gallo
- Departamento de Ciencias Celulares y Moleculares/Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Andrea Lopez Mato
- Chair of Psychoneuroimmunoendocrinology, Maimonides University, Buenos Aires, Argentina
| | - Florence Thibaut
- Department of Psychiatry, University Hospital Cochin-Site Tarnier 89 rue d’Assas, INSERM 894, Faculty of Medicine Paris Descartes, Paris, France
| | - Simone Lista
- AXA Research Fund & UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
| | - Daniel Alcolea
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Spain
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | | | |
Collapse
|
6
|
Electroconvulsive therapy does not alter the synaptic protein neurogranin in the cerebrospinal fluid of patients with major depression. J Neural Transm (Vienna) 2017; 124:1641-1645. [PMID: 29063349 DOI: 10.1007/s00702-017-1802-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 10/16/2017] [Indexed: 01/25/2023]
Abstract
Neurogranin (Ng) is a dendritic protein associated with synaptic plasticity, proposed to be a novel biomarker to measure synaptic dysfunction and degeneration in Alzheimer's disease. Since electroconvulsive therapy (ECT) has been suggested to facilitate neurogenesis and neural plasticity, we tested whether ECT could modify CSF Ng concentrations measured before and after a course of ECT in 12 patients with major depression. CSF Ng concentrations did not change, but baseline levels were positively correlated with the therapeutic response.
Collapse
|
7
|
Abstract
A set of core cerebrospinal fluid (CSF) biomarkers for Alzheimer's disease (AD) includes total tau (T-tau), phosphorylated tau (P-tau) and β-amyloid 42 (Aβ42). These biomarkers reflect some of the key aspects of AD pathophysiology, including neuronal degeneration, tau phosphorylation with tangle formation, and Aβ aggregation with deposition of the peptide into plaques. The core AD CSF biomarkers have been validated clinically in numerous studies, and found to have a very high diagnostic performance to identify AD, both in the dementia and in the mild cognitive impairment stages of the disease. CSF Aβ42 has also been found to show very high concordance with amyloid PET to identify brain amyloid deposition. The synaptic protein neurogranin is a novel candidate CSF biomarker for AD and prodromal AD. High CSF neurogranin predicts future cognitive decline and seems to be more specific for AD than, for example, T-tau. Importantly, technical developments have given ultrasensitive measurement techniques that allow measurement of brain-specific proteins such as tau and neurofilament light (NFL) in blood samples. Both plasma tau and NFL are increased in AD, and a recent study showed that plasma NFL has a diagnostic performance comparable to the core AD CSF biomarkers, and predicted future cognitive decline. Future large longitudinal clinical studies are warranted to determine the potential for plasma tau and NFL to serve as first-in-line screening tools for neurodegeneration in primary care.
Collapse
Affiliation(s)
- Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
| |
Collapse
|
8
|
Shin SS, Bales JW, Yan HQ, Kline AE, Wagner AK, Lyons-Weiler J, Dixon CE. The effect of environmental enrichment on substantia nigra gene expression after traumatic brain injury in rats. J Neurotrauma 2013; 30:259-70. [PMID: 23094804 DOI: 10.1089/neu.2012.2462] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Experimental investigations into the effects of traumatic brain injury (TBI) have demonstrated significant alterations in dopaminergic systems. Dopaminergic fibers originating within the substantia nigra and ventral tegmental area (VTA) are important for reward learning, addiction, movement, and behavior. However, little is known about the effect of TBI on substantia nigra and VTA function. Environmental enrichment (EE) has been shown to improve functional outcome after TBI, and a number of studies suggest that it may exert some benefits via dopaminergic signaling. To better understand the role of dopamine in chronic TBI pathophysiology and the effect of EE, we examined the mRNA expression profile within the substantia nigra and VTA at 4 weeks post-injury. Specifically, three comparisons were made: 1) TBI versus sham, 2) sham+EE versus sham+standard (STD) housing, and 3) TBI+EE versus TBI+STD. There were differential expressions of 25, 4, and 40 genes in these comparisons, respectively. Chronic alterations in genes post-injury within the substantia nigra and VTA included genes important for cellular membrane homeostasis and transcription. EE-induced gene alterations after TBI included genes important for signal transduction, in particular calcium signaling pathways, membrane homeostasis, and metabolism. Elucidation of these alterations in gene expression within the substantia nigra and VTA provides new insights into chronic changes in dopamine signaling post-TBI, and the potential role of EE in TBI rehabilitation.
Collapse
Affiliation(s)
- Samuel S Shin
- Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Aroclor 1254, a developmental neurotoxicant, alters energy metabolism- and intracellular signaling-associated protein networks in rat cerebellum and hippocampus. Toxicol Appl Pharmacol 2011; 256:290-9. [PMID: 21791222 DOI: 10.1016/j.taap.2011.07.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 07/06/2011] [Accepted: 07/08/2011] [Indexed: 01/23/2023]
Abstract
The vast literature on the mode of action of polychlorinated biphenyls (PCBs) indicates that PCBs are a unique model for understanding the mechanisms of toxicity of environmental mixtures of persistent chemicals. PCBs have been shown to adversely affect psychomotor function and learning and memory in humans. Although the molecular mechanisms for PCB effects are unclear, several studies indicate that the disruption of Ca(2+)-mediated signal transduction plays significant roles in PCB-induced developmental neurotoxicity. Culminating events in signal transduction pathways include the regulation of gene and protein expression, which affects the growth and function of the nervous system. Our previous studies showed changes in gene expression related to signal transduction and neuronal growth. In this study, protein expression following developmental exposure to PCB is examined. Pregnant rats (Long Evans) were dosed with 0.0 or 6.0mg/kg/day of Aroclor-1254 from gestation day 6 through postnatal day (PND) 21, and the cerebellum and hippocampus from PND14 animals were analyzed to determine Aroclor 1254-induced differential protein expression. Two proteins were found to be differentially expressed in the cerebellum following PCB exposure while 18 proteins were differentially expressed in the hippocampus. These proteins are related to energy metabolism in mitochondria (ATP synthase, sub unit β (ATP5B), creatine kinase, and malate dehydrogenase), calcium signaling (voltage-dependent anion-selective channel protein 1 (VDAC1) and ryanodine receptor type II (RyR2)), and growth of the nervous system (dihydropyrimidinase-related protein 4 (DPYSL4), valosin-containing protein (VCP)). Results suggest that Aroclor 1254-like persistent chemicals may alter energy metabolism and intracellular signaling, which might result in developmental neurotoxicity.
Collapse
|
10
|
Massaad CA, Klann E. Reactive oxygen species in the regulation of synaptic plasticity and memory. Antioxid Redox Signal 2011; 14:2013-54. [PMID: 20649473 PMCID: PMC3078504 DOI: 10.1089/ars.2010.3208] [Citation(s) in RCA: 443] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The brain is a metabolically active organ exhibiting high oxygen consumption and robust production of reactive oxygen species (ROS). The large amounts of ROS are kept in check by an elaborate network of antioxidants, which sometimes fail and lead to neuronal oxidative stress. Thus, ROS are typically categorized as neurotoxic molecules and typically exert their detrimental effects via oxidation of essential macromolecules such as enzymes and cytoskeletal proteins. Most importantly, excessive ROS are associated with decreased performance in cognitive function. However, at physiological concentrations, ROS are involved in functional changes necessary for synaptic plasticity and hence, for normal cognitive function. The fine line of role reversal of ROS from good molecules to bad molecules is far from being fully understood. This review focuses on identifying the multiple sources of ROS in the mammalian nervous system and on presenting evidence for the critical and essential role of ROS in synaptic plasticity and memory. The review also shows that the inability to restrain either age- or pathology-related increases in ROS levels leads to opposite, detrimental effects that are involved in impairments in synaptic plasticity and memory function.
Collapse
Affiliation(s)
- Cynthia A Massaad
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA.
| | | |
Collapse
|
11
|
Díez-Guerra FJ. Neurogranin, a link between calcium/calmodulin and protein kinase C signaling in synaptic plasticity. IUBMB Life 2010; 62:597-606. [DOI: 10.1002/iub.357] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
12
|
Regulation of protein kinase C isozymes during early postnatal hippocampal development. Brain Res 2009; 1288:29-41. [PMID: 19591813 DOI: 10.1016/j.brainres.2009.06.074] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Revised: 05/29/2009] [Accepted: 06/23/2009] [Indexed: 11/22/2022]
Abstract
During neonatal hippocampal development, serotonin 1A receptor-mediated signaling initially employs PKCepsilon to boost neuronal proliferation and then uses PKCalpha to promote synaptogenesis. Such stage-specific involvement of a PKC isozyme could be determined by its relative expression level. In mouse hippocampi, we detected relatively low levels of alpha, beta, gamma, and delta isozymes at postnatal days 2-6 (P2-6), which was followed by a large increase in their expression. In contrast, the PKC isozymes epsilon and theta were relatively abundant at P6, following which they underwent a further increase by P15. Comparison with purified proteins confirmed that the PKCepsilon levels at P6 and P15 were respectively 1.75 and 7.36 ng per 60 microg of protein, whereas PKCalpha levels at P6 and P15 were respectively 160 pg and 1.186 ng per 60 microg of protein. Therefore, at P6, PKCepsilon was about 11-fold more abundant than PKCalpha. Consequently, signaling cascades could use the relatively abundant PKCepsilon (and possibly PKCtheta) molecules for early events at P2-6 (e.g. neurogenesis), following which PKCalpha (and the beta, gamma, or delta isozymes) could guide maturation or apoptosis. Notably, at P6 but not P15, PKCepsilon, was localized to the nuclei of neuroblasts, probably directing mitosis. In contrast, at P15 but not P6, PKCalpha was highly expressed in the processes of the differentiated hippocampal neurons. In summary, PKC isozymes follow differential profiles of expression in neonatal hippocampus and the relative abundance of each may determine its mode and stage of involvement in hippocampal development.
Collapse
|
13
|
Vallortigara J, Chassande O, Higueret P, Enderlin V. Thyroid hormone receptor alpha plays an essential role in the normalisation of adult-onset hypothyroidism-related hypoexpression of synaptic plasticity target genes in striatum. J Neuroendocrinol 2009; 21:49-56. [PMID: 19094093 DOI: 10.1111/j.1365-2826.2008.01802.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Thyroid hormone (TH) deficiency leads to molecular changes resulting in behavioural deficits. TH action is mediated by two types of nuclear receptors (TRs), TRalpha and TRbeta, which control target gene transcription. The relative contributions of the two TR products in mediating adult TH responses are poorly understood. As TRalpha1 transcripts are widely distributed in the brain, they presumably mediate most of the TH effects. This report examines the role and specific functions of T3 receptor isoforms on regulation of striatal synaptic plasticity indicators using adult hypothyroid mutant mice that fail to express single or multiple TR gene products. We then evaluated the effect of this hypothyroidism, with or without subsequent administration of T3, on T3 nuclear receptor (TRalpha1, TRbeta) and synaptic plasticity gene expression in TRalpha(0/0), TRbeta(-/-) and wild-type 129/SV mice. Hypothyroid wild-type mice exhibited reduced TRbeta, RC3, CaMKII and Rhes expression. The mRNA levels of Rhes and CaMKII were the same in all three hypothyroid substrains. By contrast, hypothyroid TRbeta(-/-) mice had higher RC3 mRNA levels than wild-type. T3 administration restored TRbeta, RC3 and CaMKII levels in hypothyroid wild-type mice, without significant Rhes upregulation. T3 administration normalised expression of all genes studied in hypothyroid TRbeta(-/-) but not TRalpha(0/0) mice. Thus, TRalpha apparently plays an essential role in restoring the expression of the TH-regulated genes potentially involved in striatal synaptic plasticity.
Collapse
Affiliation(s)
- J Vallortigara
- Unité de Nutrition et Neurosciences, Universités Bordeaux 1-Bordeaux 2, Avenue des Facultés, Talence, France
| | | | | | | |
Collapse
|
14
|
Vallortigara J, Alfos S, Micheau J, Higueret P, Enderlin V. T3 administration in adult hypothyroid mice modulates expression of proteins involved in striatal synaptic plasticity and improves motor behavior. Neurobiol Dis 2008; 31:378-85. [PMID: 18585460 DOI: 10.1016/j.nbd.2008.05.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2007] [Revised: 05/16/2008] [Accepted: 05/19/2008] [Indexed: 12/01/2022] Open
Abstract
Adult-onset hypothyroidism is associated with neurological changes such as cognitive dysfunction and impaired learning, which may be related to alterations of synaptic plasticity. We investigate the consequence of adult-onset hypothyroidism on thyroid-mediated transcription events in striatal synaptic plasticity, and the effect of triiodothyronine (T3) replacement. We used hypothyroid mice, treated with propylthiouracil (PTU) and methimazole (MMI), with or without subsequent administration of T3. We evaluated the amount of T3 nuclear receptors (TRalpha1, TRbeta) and striatal plasticity indicators: neurogranin (RC3), Ras homolog enriched in striatum (Rhes), Ca2+/calmodulin-dependent protein kinase (CaMKII), and dopamine- and cAMP-regulated phosphoprotein (DARPP-32). In addition, we assessed hypothyroid mice motor behavior as related to striatum synaptic functions. Hypothyroid mice exhibited significantly reduced TRbeta, RC3 and Rhes expression. T3 administration reversed the expression of TRbeta, RC3, and up-regulated CaMKII levels as well as motor behavior, and decreased DARPP-32 protein phosphorylation. We suggest that thyroid hormone modulation had a major impact on striatal synaptic plasticity of adult mice which produced in turn motor behavior modifications.
Collapse
Affiliation(s)
- Julie Vallortigara
- Unité de Nutrition et Neurosciences, Universités Bordeaux 1-Bordeaux 2, Avenue des Facultés, 33405 Talence Cedex, France
| | | | | | | | | |
Collapse
|
15
|
Krueger DD, Nairn AC. Expression of PKC substrate proteins, GAP-43 and neurogranin, is downregulated by cAMP signaling and alterations in synaptic activity. Eur J Neurosci 2007; 26:3043-53. [PMID: 18005072 DOI: 10.1111/j.1460-9568.2007.05901.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Growth-associated protein 43 (GAP-43) and neurogranin are protein kinase C substrate proteins that are thought to play an important role in synaptic plasticity, but little is currently known about the mechanisms that may regulate their function at the synapse. In this study, we show that long-term elevation of intracellular cAMP levels in rat primary cortical cultures results in a persistent downregulation of GAP-43 and neurogranin, most likely at the transcriptional level. This effect may be at least partially mediated by protein kinase A, but is independent of protein kinase C activation. Moreover, it is mimicked and occluded by manipulations that alter the levels of spontaneous synaptic activity in primary cultures, such as bicuculline and tetrodotoxin. These data suggest that levels of GAP-43 and neurogranin are regulated by factors known to modulate synaptic strength, thus providing a potential mechanism by which protein kinase C signaling pathways and their substrates might contribute to synaptic function and/or plasticity.
Collapse
Affiliation(s)
- Dilja D Krueger
- Department of Psychiatry, Division of Molecular Psychiatry, Yale University School of Medicine, Ribicoff Research Facilities, CMHC, 34 Park Street, New Haven, CT 06508, USA
| | | |
Collapse
|
16
|
Higo N, Oishi T, Yamashita A, Murata Y, Matsuda K, Hayashi M. Expression of protein kinase-C substrate mRNA in the motor cortex of adult and infant macaque monkeys. Brain Res 2007; 1171:30-41. [PMID: 17761152 DOI: 10.1016/j.brainres.2007.07.054] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2007] [Revised: 07/24/2007] [Accepted: 07/24/2007] [Indexed: 10/23/2022]
Abstract
To understand the molecular and cellular bases of plasticity in the primate motor cortex, we investigated the expression of three protein kinase-C (PKC) substrates: GAP-43, myristoylated alanine-rich C-kinase substrate (MARCKS), and neurogranin, which are key molecules regulating synaptic plasticity. Prominent signals for the three mRNAs were primarily observed in pyramidal cells. Large pyramidal cells in layer V, from which the descending motor tract originates, contained weaker hybridization signals for GAP-43 and neurogranin mRNAs than did the smaller pyramidal cells. We also performed double-label in situ hybridization showing that GAP-43 and neurogranin mRNAs were expressed in a subset of MARCKS-positive neurons. Quantitative analysis showed that the expression was different between the layers: layer VI contained the strongest and layer II the weakest signals for all three mRNAs. The expression levels of GAP-43 and MARCKS mRNA in layer V were higher than in layer III, while the expression level of neurogranin mRNA in layer V was almost the same as in layer III. Developmental analysis from the newborn to adult indicated that the expression levels of the three mRNAs were higher in the infant motor cortex than in the adult. The expression of both GAP-43 and neurogranin mRNAs transiently increased over several months postnatally. The present study showed that the expression of the three PKC substrates was specific to cell types, cortical layers, and postnatal developmental stage. The specific expression may reflect functional specialization for plasticity in the motor cortex of both infants and adults.
Collapse
Affiliation(s)
- Noriyuki Higo
- Neuroscience Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Umezono, Tsukuba, Ibaraki, Japan.
| | | | | | | | | | | |
Collapse
|
17
|
Coburn CG, Currás-Collazo MC, Kodavanti PRS. In vitro effects of environmentally relevant polybrominated diphenyl ether (PBDE) congeners on calcium buffering mechanisms in rat brain. Neurochem Res 2007; 33:355-64. [PMID: 17846885 DOI: 10.1007/s11064-007-9430-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2007] [Accepted: 06/29/2007] [Indexed: 11/26/2022]
Abstract
Polybrominated diphenyl ethers (PBDEs) are widely used as additive flame-retardants and have been detected in human blood, adipose tissue, and breast milk. Developmental and long-term exposures to these chemicals may pose a human health risk, especially to children. We have previously demonstrated that polychlorinated biphenyls (PCBs), which are structurally similar to PBDEs and cause neurotoxicity, perturb intracellular signaling events including calcium homeostasis and protein kinase C translocation, which are critical for neuronal function and development of the nervous system. The objective of the present study was to test whether environmentally relevant PBDE congeners 47 and 99 are also capable of disrupting Ca(2+) homeostasis. Calcium buffering was determined by measuring (45)Ca(2+)-uptake by microsomes and mitochondria, isolated from adult male rat brain (frontal cortex, cerebellum, hippocampus, and hypothalamus). Results show that PBDEs 47 and 99 inhibit both microsomal and mitochondrial (45)Ca(2+)-uptake in a concentration-dependent manner. The effect of these congeners on (45)Ca(2+)-uptake is similar in all four brain regions though the hypothalamus seems to be slightly more sensitive. Among the two preparations, the congeners inhibited (45)Ca(2+)-uptake in mitochondria to a greater extent than in microsomes. These results indicate that PBDE 47 and PBDE 99 congeners perturb calcium signaling in rat brain in a manner similar to PCB congeners, suggesting a common mode of action of these persistent organic pollutants.
Collapse
Affiliation(s)
- Cary G Coburn
- Environmental Toxicology Graduate Program, University of California, Riverside, CA 92521, USA
| | | | | |
Collapse
|
18
|
Bachmann RF, Schloesser RJ, Gould TD, Manji HK. Mood stabilizers target cellular plasticity and resilience cascades: implications for the development of novel therapeutics. Mol Neurobiol 2007; 32:173-202. [PMID: 16215281 DOI: 10.1385/mn:32:2:173] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Bipolar disorder is a devastating disease with a lifetime incidence of about 1% in the general population. Suicide is the cause of death in 10 to 15% of patients and in addition to suicide, mood disorders are associated with many other harmful health effects. Mood stabilizers are medications used to treat bipolar disorder. In addition to their therapeutic effects for the treatment of acute manic episodes, mood stabilizers are useful as prophylaxis against future episodes and as adjunctive antidepressant medications. The most established and investigated mood-stabilizing drugs are lithium and valproate but other anticonvulsants (such as carbamazepine and lamotrigine) and antipsychotics are also considered as mood stabilizers. Despite the efficacy of these diverse medications, their mechanisms of action remain, to a great extent, unknown. Lithium's inhibition of some enzymes, such as inositol monophosphatase and glycogen synthase kinase-3, probably results in its mood-stabilizing effects. Valproate may share its anticonvulsant target with its mood-stabilizing target or may act through other mechanisms. It has been shown that lithium, valproate, and/or carbamazepine regulate numerous factors involved in cell survival pathways, including cyclic adenine monophospate response element-binding protein, brain-derived neurotrophic factor, bcl-2, and mitogen-activated protein kinases. These drugs have been suggested to have neurotrophic and neuroprotective properties that ameliorate impairments of cellular plasticity and resilience underlying the pathophysiology of mood disorders. This article also discusses approaches to develop novel treatments specifically for bipolar disorder.
Collapse
Affiliation(s)
- Rosilla F Bachmann
- Laboratory of Molecular Pathophysiology, National Institute of Mental Health, Bethesda, MD, USA
| | | | | | | |
Collapse
|
19
|
Han NLR, Wen J, Lin Q, Tan PL, Liou YC, Sheu FS. Proteomics analysis of the expression of neurogranin in murine neuroblastoma (Neuro-2a) cells reveals its involvement for cell differentiation. Int J Biol Sci 2007; 3:263-73. [PMID: 17505539 PMCID: PMC1865092 DOI: 10.7150/ijbs.3.263] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2007] [Accepted: 04/17/2007] [Indexed: 11/05/2022] Open
Abstract
Neurogranin (Ng) is a neural-specific, calmodulin (CaM)-binding protein that is phosphorylated by protein kinase C (PKC). Although its biochemical property has been well characterized, the physiological function of Ng needs to be elucidated. In the present study, we performed proteomics analysis of the induced compositional changes due to the expression of Ng in murine neuroblastoma (Neuro-2a) cells using isotope coded affinity tags (ICAT) combined with 2-dimensional liquid chromatography/tandem mass spectrometry (2D-LC/MS/MS). We found that 40% of identified proteins were down-regulated and most of these proteins are microtubule components and associated proteins that mediated neurite outgrowth. Western blot experiments confirmed the expression of α-tubulin and microtubule- associated protein 1B (MAP 1B) was dramatically reduced in Neuro-2a-Ng cells compared to control. Cell morphology of Neuro-2a-Ng showed far less neurites than the control. Serum deprivation induced the extension of only one or two long neurites per cell in Neuro-2a-Ng, contrasting to the extension of multiple neurites per control cell. Ng may be linked to neurite formation by affecting expression of several microtubule related proteins. Furthermore, the PKC activator (PMA) induced an enhanced ERK1/2 activity in the cells that expressed Ng. The mutation of Ng at S36A caused sustained increase of ERK1/2 activity, whereas the ERK1/2 activity in mutation at I33Q showed no difference compared to wild type Ng, suggesting the phosphorylation of Ng but not the CaM /Ng interaction plays an important role in ERK activation. Ng may be involved in neuronal growth and differentiation via PKC and ERK1/2 signaling pathways.
Collapse
Affiliation(s)
- Nian-Lin Reena Han
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | | | | | | | | | | |
Collapse
|
20
|
Abstract
A number of neuronal functions, including synaptic plasticity, depend on proper regulation of synaptic proteins, many of which can be rapidly regulated by phosphorylation. Neuronal activity controls the function of these synaptic proteins by exquisitely regulating the balance of various protein kinase and protein phosphatase activity. Recent understanding of synaptic plasticity mechanisms underscores important roles that these synaptic phosphoproteins play in regulating both pre- and post-synaptic functions. This review will focus on key postsynaptic phosphoproteins that have been implicated to play a role in synaptic plasticity.
Collapse
Affiliation(s)
- Hey-Kyoung Lee
- Department of Biology, Neuroscience and Cognitive Science (NACS) Program, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
21
|
Kodavanti PRS. Neurotoxicity of persistent organic pollutants: possible mode(s) of action and further considerations. Dose Response 2006; 3:273-305. [PMID: 18648619 PMCID: PMC2475949 DOI: 10.2203/dose-response.003.03.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Persistent organic pollutants (POPs) are long-lived toxic organic compounds and are of major concern for human and ecosystem health. Although the use of most POPs is banned in most countries, some organochlorine pesticides are still being used in several parts of the world. Although environmental levels of some POPs such as polychlorinated biphenyls (PCBs) have declined, newly emerging POPs such as polybrominated diphenyl ethers (PBDEs) have been increasing considerably. Exposure to POPs has been associated with a wide spectrum of effects including reproductive, developmental, immunologic, carcinogenic, and neurotoxic effects. It is of particular concern that neurotoxic effects of some POPs have been observed in humans at low environmental concentrations. This review focuses on PCBs as a representative chemical class of POPs and discusses the possible mode(s) of action for the neurotoxic effects with emphasis on comparing dose-response and structure-activity relationships (SAR) with other structurally related chemicals. There is sufficient epidemiological and experimental evidence showing that PCB exposure is associated with motor and cognitive deficits in humans and animal models. Although several potential mode(s) of actions were postulated for PCB-induced neurotoxic effects, changes in neurotransmitter systems, altered intracellular signalling processes, and thyroid hormone imbalance are predominant ones. These three potential mechanisms are discussed in detail in vitro and in vivo. In addition, SAR was conducted on other structurally similar chemicals to see if they have a common mode(s) of action. Relative potency factors for several of these POPs were calculated based on their effects on intracellular signalling processes. This is a comprehensive review comparing molecular effects at the cellular level to the neurotoxic effects seen in the whole animal for environmentally relevant POPs.
Collapse
Affiliation(s)
- Prasada Rao S Kodavanti
- Cellular and Molecular Toxicology Branch, Neurotoxicology Division, NHEERL/ORD, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| |
Collapse
|
22
|
Hori K, Yasuda H, Konno D, Maruoka H, Tsumoto T, Sobue K. NMDA receptor-dependent synaptic translocation of insulin receptor substrate p53 via protein kinase C signaling. J Neurosci 2006; 25:2670-81. [PMID: 15758177 PMCID: PMC6725157 DOI: 10.1523/jneurosci.3638-04.2005] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The activity-dependent remodeling of postsynaptic structure is a fundamental process underlying learning and memory. Insulin receptor substrate p53 (IRSp53), a key player in cytoskeletal dynamics, is enriched in the postsynaptic density (PSD) fraction, but its significance in synaptic functions remains unclear. We report here that IRSp53 is accumulated rapidly at the postsynaptic sites of cultured hippocampal neurons after glutamate or NMDA stimulation in an actin cytoskeleton-dependent manner. Pharmacological profiles showed that a PKC inhibitor, but not other kinase inhibitors, specifically suppressed the synaptic translocation of IRSp53 in response to NMDA, and the selective activation of PKC with phorbol ester markedly induced the synaptic translocation. Reverse transcriptase-PCR and Western blotting showed that IRSp53-S is the major isoform expressed in cultured hippocampal neurons. The synaptic targeting of IRSp53-S was found to be mediated through N-terminal coiled-coil domain and the PDZ (PSD-95/Discs large/zona occludens-1)-binding sequence at its C-terminal end and regulated by the PKC phosphorylation of its N terminus. In electrophysiological experiments, overexpression of IRSp53-S wild type and IRSp53-S mutant that is spontaneously accumulated at the postsynaptic sites enhanced the postsynaptic function as detected by an increased miniature EPSC amplitude. These data suggest that IRSp53 is involved in NMDA receptor-linked synaptic plasticity via PKC signaling.
Collapse
Affiliation(s)
- Kei Hori
- Department of Neuroscience (D13) and Division of Neurophysiology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | | | | | | | | | | |
Collapse
|
23
|
Higo N, Oishi T, Yamashita A, Murata Y, Matsuda K, Hayashi M. Northern blot and in situ hybridization analyses for the neurogranin mRNA in the developing monkey cerebral cortex. Brain Res 2006; 1078:35-48. [PMID: 16497282 DOI: 10.1016/j.brainres.2006.01.062] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2005] [Revised: 01/03/2006] [Accepted: 01/08/2006] [Indexed: 11/22/2022]
Abstract
Neurogranin is a postsynaptic substrate for protein kinase C, and its expression is related to dendritic spine development and postsynaptic plasticity. Using both Northern blot analysis and in situ hybridization techniques, we investigated the developmental changes of neurogranin expression in the monkey cerebral cortex. In each of four neocortical areas examined, i.e., the prefrontal area (area FD of von Bonin and Bailey), the temporal association area (TE), the primary somatosensory area (PB), and the primary visual area (OC), the Northern blot analysis showed that the amount of neurogranin mRNA was low during the prenatal and perinatal periods until postnatal day 8. It increased during postnatal development and reached its peak value at postnatal day 70 (in area OC) or postnatal month 6 (in area FD, TE, and PB). After that, the amount of neurogranin mRNA in the cerebral neocortex decreased gradually until postnatal years 2-3. The in situ hybridization experiments also showed a transient increase of neurogranin mRNA in the neocortex during postnatal day 70 to postnatal month 6. The transient increase was prominent in layers II and III of areas FD and TE; deep in layer III of area PB; and in layers II, III, and IV of area OC. In the hippocampus, in contrast to the results in the neocortex, the expression of neurogranin mRNA was decreased almost continuously during the postnatal period. The transiently increased expression of neurogranin in the postnatal neocortex may be a molecular basis for the postsynaptic modification of afferent inputs possibly from subcortical structures.
Collapse
Affiliation(s)
- Noriyuki Higo
- Neuroscience Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Umezono, Tsukuba, Ibaraki 305-8568, Japan.
| | | | | | | | | | | |
Collapse
|
24
|
Higo N, Oishi T, Yamashita A, Murata Y, Matsuda K, Hayashi M. Expression of protein kinase C-substrate mRNAs in the basal ganglia of adult and infant macaque monkeys. J Comp Neurol 2006; 499:662-76. [PMID: 17029258 DOI: 10.1002/cne.21119] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We performed in situ hybridization histochemistry on the monkey basal ganglia to investigate the mRNA localization of three protein kinase C substrates (GAP-43, MARCKS, and neurogranin), of which expression plays a role in structural changes in neurites and synapses. Weak hybridization signals for GAP-43 mRNA and intense signals for both MARCKS and neurogranin mRNAs were observed in the adult neostriatum. All three of the mRNAs were expressed in both substance P-positive direct pathway neurons and enkephalin-positive indirect pathway neurons. In the nucleus accumbens, the hybridization signals for the three mRNAs were weaker than those in the neostriatum. Double-label in situ hybridization histochemistry in the neostriatum revealed that GAP-43 and neurogranin mRNAs were expressed in a subset of MARCKS-positive neurons. While intense hybridization signals for MARCKS mRNA were observed in all of the other basal ganglia regions such as the globus pallidus, substantia innominata, subthalamic nucleus, and substantia nigra, intense signals for GAP-43 mRNA were restricted to the substantia innominata and substantia nigra pars compacta. No signal for neurogranin mRNA was observed in the basal ganglia regions outside the neostriatum and the nucleus accumbens. These results indicate that the protein kinase C substrates are abundant in some specific connections in cortico-basal ganglia circuits. Developmental analysis showed that the expression level in the putamen and nucleus accumbens, but not in the caudate nucleus, was higher in the infant than in the adult, suggesting that synaptic maturation in the caudate nucleus occurs earlier than that in the putamen and nucleus accumbens.
Collapse
Affiliation(s)
- Noriyuki Higo
- Neuroscience Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki 305-8568, Japan.
| | | | | | | | | | | |
Collapse
|
25
|
Murata Y, Higo N, Oishi T, Yamashita A, Matsuda K, Hayashi M. Developmental changes in the expression of growth-associated protein-43 mRNA in the monkey thalamus: northern blot and in situ hybridization studies. Neuroscience 2005; 136:497-507. [PMID: 16203103 DOI: 10.1016/j.neuroscience.2005.08.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2005] [Revised: 07/06/2005] [Accepted: 08/04/2005] [Indexed: 12/01/2022]
Abstract
The expression of growth-associated protein-43 has been related to axonal elongation and synaptic sprouting. Using the Northern blot analysis, we investigated the developmental changes of growth-associated protein-43 mRNA in the thalamus of macaque monkeys. The amount of growth-associated protein-43 mRNA was high at embryonic day 125, and decreased at postnatal day 1. It increased again at postnatal day 8, reached its peak value at postnatal days 50-70, and then decreased gradually until postnatal year 1. We previously reported that the amount of growth-associated protein-43 mRNA in the cerebral cortex decreased roughly exponentially during perinatal and postnatal periods and that it approached the asymptote by postnatal day 70 [Oishi T, Higo N, Umino Y, Matsuda K, Hayashi M (1998) Development of GAP-43 mRNA in the macaque cerebral cortex. Dev Brain Res 109:87-97]. The present findings may indicate that extensive synaptic growth of thalamic neurons continues even after that of cortical neurons has finished. We then performed in situ hybridization to investigate whether the expression level of growth-associated protein-43 mRNA was different among various thalamic nuclei. In the infant thalamus (postnatal days 70-90), moderate to intense expression of growth-associated protein-43 mRNA was detected in all thalamic nuclei. Quantitative analysis in the infant thalamus indicated that the expression levels were different between the nuclear groups that are defined by the origin of their afferents. The expression in the first order nuclei, which receive their primary afferent fibers from ascending pathways [Guillery RW (1995) Anatomical evidence concerning the role of the thalamus in corticocortical communication: a brief review. J Anat 187 (Pt 3):583-592], was significantly higher than that in the higher order nuclei. While moderate expression was also detected in the adult dorsal thalamus, the expression in the first order nuclei was almost the same as that in the higher order nuclei. Thus, the in situ hybridization experiments indicated that the transient postnatal increase in the amount of growth-associated protein-43 mRNA, which was shown by the Northern blot analysis, was mainly attributed to enhanced expression in the first order nuclei during the postnatal period. This may be a molecular basis for environmentally induced modification of thalamocortical synapses.
Collapse
Affiliation(s)
- Y Murata
- Neuroscience Research Institute, National Institute of Advanced Industrial Science and Technology, Umezono, Tsukuba, Ibaraki 305-8568, Japan
| | | | | | | | | | | |
Collapse
|
26
|
Féart C, Mingaud F, Enderlin V, Husson M, Alfos S, Higueret P, Pallet V. Differential effect of retinoic acid and triiodothyronine on the age-related hypo-expression of neurogranin in rat. Neurobiol Aging 2005; 26:729-38. [PMID: 15708448 DOI: 10.1016/j.neurobiolaging.2004.06.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2004] [Revised: 06/10/2004] [Accepted: 06/21/2004] [Indexed: 12/20/2022]
Abstract
Given the important role of retinoids and thyroid hormone for optimal brain functioning and the tenuous relationship between retinoic acid (RA) and triiodothyronine (T3) signalings, we compared the effects of RA or T3 administrations on RA and T3 nuclear receptors (RAR, RXR and TR) and on their target genes, neuromodulin (GAP43) and neurogranin (RC3) in 24-month-old rats. Quantitative real time PCR and western blot analysis allowed us to verify that retinoid and thyroid signalings and GAP43 and RC3 expression are affected by age. By in situ hybridization we observed a decreased expression of RC3 in hippocampus, striatum and cerebral cortex. RARbeta, RXRbeta/gamma and GAP43 were up-regulated by RA as well as T3 treatment. The abundance of TRalpha/beta mRNA and RC3 expression were only increased by T3 administration in the whole brain. This up-regulator effect of T3 on RC3 was only observed in the striatum. During aging, T3 become a limiting factor alone able to correct the age-related concomitant hypo-activation of retinoid and thyroid signalings and alterations of synaptic plasticity.
Collapse
Affiliation(s)
- C Féart
- Unité de Nutrition et Signalisation Cellulaire (E.A. MENRT; USC INRA) ISTAB, Avenue des Facultés, Université Bordeaux I, 33405 Talence cedex, France
| | | | | | | | | | | | | |
Collapse
|
27
|
Kodavanti PRS, Ward TR. Differential Effects of Commercial Polybrominated Diphenyl Ether and Polychlorinated Biphenyl Mixtures on Intracellular Signaling in Rat Brain in Vitro. Toxicol Sci 2005; 85:952-62. [PMID: 15772365 DOI: 10.1093/toxsci/kfi147] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) are widely used as flame retardants and have been detected in human blood, adipose tissue, and breast milk. Developmental and long-term exposures to these contaminants may pose a human health risk, especially to children. Previously, we demonstrated that polychlorinated biphenyls (PCBs), which are neurotoxic and structurally similar to PBDEs, perturbed intracellular signaling events, including calcium homeostasis and subsequent events such as protein kinase C (PKC), which are critical for the normal function and development of the nervous system. The objective of the present study was to test whether commercial PBDE mixtures (DE-71, a pentabrominated dipheyl ether mixture, and DE-79, a mostly octabromodiphenyl ether mixture) affected intracellular signaling mechanisms in a similar way to that of PCBs and other organohalogens, as an attempt to understand the common mode of action for these persistent chemicals. PKC translocation was studied by determining (3)H-phorbol ester ((3)H-PDBu) binding in rat cerebellar granule cells, and calcium buffering was determined by measuring (45)Ca(2+) uptake by microsomes and mitochondria isolated from adult male rat brain (frontal cortex, cerebellum, and hippocampus). As seen with PCBs, DE-71 increased PKC translocation and inhibited (45)Ca(2+) uptake by both microsomes and mitochondria in a concentration-dependent manner. The effect of DE-71 on (45)Ca(2+) uptake seems to be similar in all three brain regions. Between the two organelles, DE-71 inhibited mitochondrial (45)Ca(2+) uptake to a greater extent than microsomal (45)Ca(2+) uptake. DE-79 had no effects on either neurochemical event even at 30 mug/ml. Aroclor 1254 altered both events to a greater extent compared to DE-71 on a weight basis. When the results were compared on a molar basis, Aroclor 1254 altered PKC translocation and microsomal (45)CaP(2+) uptake to a greater extent than DE-71, however, Aroclor 1254 and DE-71 equally affected mitochondrial (45)Ca(2+) uptake. These results indicate that PBDEs perturbed intracellular signaling mechanisms in rat brain as do other organohalogen compounds and the efficacy between the commercial PCB and PBDE mixtures seem to vary with different endpoints.
Collapse
Affiliation(s)
- Prasada Rao S Kodavanti
- Cellular and Molecular Toxicology Branch, Neurotoxicology Division, NHEERL, ORD, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA.
| | | |
Collapse
|
28
|
Ethell IM, Pasquale EB. Molecular mechanisms of dendritic spine development and remodeling. Prog Neurobiol 2005; 75:161-205. [PMID: 15882774 DOI: 10.1016/j.pneurobio.2005.02.003] [Citation(s) in RCA: 265] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2004] [Revised: 01/28/2005] [Accepted: 02/22/2005] [Indexed: 12/19/2022]
Abstract
Dendritic spines are small protrusions that cover the surface of dendrites and bear the postsynaptic component of excitatory synapses. Having an enlarged head connected to the dendrite by a narrow neck, dendritic spines provide a postsynaptic biochemical compartment that separates the synaptic space from the dendritic shaft and allows each spine to function as a partially independent unit. Spines develop around the time of synaptogenesis and are dynamic structures that continue to undergo remodeling over time. Changes in spine morphology and density influence the properties of neural circuits. Our knowledge of the structure and function of dendritic spines has progressed significantly since their discovery over a century ago, but many uncertainties still remain. For example, several different models have been put forth outlining the sequence of events that lead to the genesis of a spine. Although spines are small and apparently simple organelles with a cytoskeleton mainly composed of actin filaments, regulation of their morphology and physiology appears to be quite sophisticated. A multitude of molecules have been implicated in dendritic spine development and remodeling, suggesting that intricate networks of interconnected signaling pathways converge to regulate actin dynamics in spines. This complexity is not surprising, given the likely importance of dendritic spines in higher brain functions. In this review, we discuss the molecules that are currently known to mediate the exquisite sensitivity of spines to perturbations in their environment and we outline how these molecules interface with each other to mediate cascades of signals flowing from the spine surface to the actin cytoskeleton.
Collapse
Affiliation(s)
- Iryna M Ethell
- Division of Biomedical Sciences, University of California Riverside, Riverside, CA 92521, USA
| | | |
Collapse
|
29
|
Singec I, Knoth R, Ditter M, Volk B, Frotscher M. Neurogranin is expressed by principal cells but not interneurons in the rodent and monkey neocortex and hippocampus. J Comp Neurol 2004; 479:30-42. [PMID: 15389613 DOI: 10.1002/cne.20302] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
As a substrate of protein kinase C (PKC), neurogranin (NG) is involved in the regulation of calcium signaling and activity-dependent plasticity. Recently, we have shown that, in the rodent cerebellum, NG is exclusively expressed by gamma-aminobutyric acidergic Golgi cells, whereas, in the monkey cerebellum, brush cells were the only neuronal population expressing NG (Singec et al. [2003] J. Comp. Neurol. 459:278-289). In the present study, we analyzed the neocortical and hippocampal expression patterns of NG in adult mouse (C57Bl/6), rat (Wistar), and monkey (Cercopithecus aetiops). By using immunocytochemistry and nonradioactive in situ hybridization, we demonstrate strong NG expression by principal cells in different neocortical layers and in the hippocampus by granule cells of the dentate gyrus and pyramidal neurons of CA1-CA3. In contrast, double-labeling experiments in rodents revealed that neocortical and hippocampal interneurons expressing glutamate decarboxylase 67 (GAD67) were consistently devoid of NG. In addition, by using antibodies against parvalbumin, calbindin, and calretinin, we could demonstrate the absence of NG in interneurons of monkey frontal cortex and hippocampus. Together these findings corroborate the idea of different calcium signaling pathways in excitatory and inhibitory cells that may contribute to different modes of synaptic plasticity in these neurons.
Collapse
Affiliation(s)
- Ilyas Singec
- Institute of Anatomy and Cell Biology, University of Freiburg, D-79104 Freiburg, Germany.
| | | | | | | | | |
Collapse
|
30
|
Husson M, Enderlin V, Alfos S, Boucheron C, Pallet V, Higueret P. Expression of neurogranin and neuromodulin is affected in the striatum of vitamin A-deprived rats. ACTA ACUST UNITED AC 2004; 123:7-17. [PMID: 15046861 DOI: 10.1016/j.molbrainres.2003.12.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2003] [Indexed: 11/15/2022]
Abstract
Our previous data showed that vitamin A deficiency (VAD) induces, in whole brain, a reduced amount of mRNA for brain retinoic acid (RA) and triiodothyronine (T3) nuclear receptors (i.e., RAR, RXR, and TR, respectively), which is accompanied by reduced amounts of mRNA and protein of neurogranin (RC3, a neuronal protein involved in synaptic plasticity) as well as selective behavioral impairment. Given the important role of retinoids for optimal brain functioning, the effects of vitamin A depletion and subsequent administration of RA or T3 on the mRNA levels of RA and T3 nuclear receptors and on two target genes' (RC3 and neuromodulin or GAP43) mRNA and protein levels were examined in the hippocampus, striatum, and cerebral cortex. A quantitative real-time polymerase chain reaction (PCR), in situ hybridization, and Western blot analysis demonstrated that the striatal region is the brain site where both RA and T3 signaling pathways are most affected by VAD. Indeed, rats fed a vitamin A-free diet for 10 weeks exhibited decreased expression of RAR, RXR, TR, RC3, and GAP43 in the striatum. The administration of T3 to these vitamin A-deprived rats reversed the reduction in mRNA levels of RA and T3 nuclear receptors and in mRNA and protein levels of target genes in this region. These data suggest that modifications that appear preferentially in the striatum, a region highly sensitive to vitamin A bioavailability, may contribute to neurobiological alterations and the spatial learning impairment that occurs in vitamin A-deprived animals.
Collapse
Affiliation(s)
- M Husson
- Unité de Nutrition et Signalisation Cellulaire (EA MENRT; USC INRA) ISTAB, Université Bordeaux 1, Avenue des Facultés, Talence Cedex 33405, France
| | | | | | | | | | | |
Collapse
|
31
|
Etchamendy N, Enderlin V, Marighetto A, Pallet V, Higueret P, Jaffard R. Vitamin A deficiency and relational memory deficit in adult mice: relationships with changes in brain retinoid signalling. Behav Brain Res 2003; 145:37-49. [PMID: 14529804 DOI: 10.1016/s0166-4328(03)00099-8] [Citation(s) in RCA: 142] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Vitamin A and its derivatives, the retinoids, have recently been reported to be implicated in the synaptic plasticity of the hippocampus and in cognitive functions. Acting via transcription factors, retinoids can regulate gene expression via their nuclear receptors [retinoic acid receptors (RARs) and retinoid X receptors (RXRs)]. We recently showed that a moderate (about 30%) hypoexpression of brain (and hippocampal) retinoid signalling, like that naturally occurring in the aged brain of mice, might be related to a selective relational memory deficit. To further assess this hypothesis, the present study investigated the effects of Vitamin A deprivation of varying duration both on the brain expression of retinoid receptors (RARbeta and RXRbeta/gamma) and two associated target genes [tissue-type transglutaminase (tTG) and neurogranin, (RC3)], and on radial maze discrimination learning using young adult mice as subjects. We observed that irrespective of its duration (i.e. 31 or 39 weeks), Vitamin A deprivation resulted in a significant reduction (25-30%) in the expression of brain RARbeta, RXRbeta/gamma and tTG mRNAs. Conversely, only the 39-week condition was found to induce a significant decrease in brain RC3 mRNAs contents and a selective relational memory impairment. Finally, daily administration of retinoic acid (RA) failed to reverse the 39-week Vitamin A deficiency (VAD)-related cognitive deficit and to fully normalise the associated brain retinoid hyposignalling. In particular, there was no evidence for an up-regulating effect of RA on whole brain (and hippocampal) RC3 mRNAs of the 39-week-depleted mice. The results show that post-natal VAD may induce a selective memory impairment and give further support to the hypothesis that the fine regulation of retinoid-mediated gene expression is important for optimal brain functioning and higher cognition.
Collapse
Affiliation(s)
- Nicole Etchamendy
- CNRS UMR 5106, Lab. Neurosciences Cognitives, Université de Bordeaux 1, Avenue des Facultés, 33405 Talence Cedex, France.
| | | | | | | | | | | |
Collapse
|
32
|
Abstract
1. It is not completely clear how volatile anesthetics cause anesthesia, but one possible consequence of their action is to alter presynaptic activity and the release of neurotransmitters due to alterations in intracellular signaling. 2. Protein kinase C (PKC) is a signal transducing enzyme that is an important regulator of multiple physiological processes like neurotransmitter release, ion channel activity, and neurotransmitter receptor desensitization. Thus, PKC is an attractive molecular target for the synaptic action of general anesthetics. 3. However, the effects of these agents on PKC activity are not yet fully understood and there are several contradictory data on the literature regarding the in vitro and in vivo preparations. 4. Here, we will review some evidence for volatile anesthetics effects on neuronal PKC activation.
Collapse
Affiliation(s)
- Renato Santiago Gomez
- Departamento de Cirurgia, Faculdade de Medicina da UFMG, Belo Horizonte-Minas Gerais, Brazil
| | | | - Marcus Vinicius Gomez
- Laboratório de Neurofarmacologia, Departamento de Farmacologia, ICB-UFMG, Belo Horizonte-Minas Gerais, Brazil
| |
Collapse
|
33
|
Higo N, Oishi T, Yamashita A, Matsuda K, Hayashi M. Cell type- and region-specific expression of protein kinase C-substrate mRNAs in the cerebellum of the macaque monkey. J Comp Neurol 2003; 467:135-49. [PMID: 14595765 DOI: 10.1002/cne.10850] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
We performed nonradioactive in situ hybridization histochemistry in the monkey cerebellum to investigate the localization of protein kinase C-substrate (growth-associated protein-43 [GAP-43], myristoylated alanine-rich C-kinase substrate [MARCKS], and neurogranin) mRNAs. Hybridization signals for GAP-43 mRNA were observed in the molecular and granule cell layers of both infant and adult cerebellar cortices. Signals for MARCKS mRNA were observed in the molecular, Purkinje cell, and granule cell layers of both infant and adult cortices. Moreover, both GAP-43 and MARCKS mRNAs were expressed in the external granule cell layer of the infant cortex. In the adult cerebellar vermis, signals for both GAP-43 and MARCKS mRNAs were more intense in lobules I, IX, and X than in the remaining lobules. In the adult hemisphere, both mRNAs were more intense in the flocculus and the dorsal paraflocculus than in other lobules. Such lobule-specific expressions were not prominent in the infant cerebellar cortex. Signals for neurogranin, a postsynaptic substrate for protein kinase C, were weak or not detectable in any regions of either the infant or adult cerebellar cortex. The prominent signals for MARCKS mRNA were observed in the deep cerebellar nuclei, but signals for both GAP-43 and neurogranin mRNAs were weak or not detectable. The prominent signals for both GAP-43 and MARCKS mRNAs were observed in the inferior olive, but signals for neurogranin were weak or not detectable. The cell type- and region-specific expression of GAP-43 and MARCKS mRNAs in the cerebellum may be related to functional specialization regarding plasticity in each type of cell and each region of the cerebellum.
Collapse
Affiliation(s)
- Noriyuki Higo
- Neuroscience Research Institute, National Institute of Advanced Industrial Science and Technology, Umezono, Tsukuba, Ibaraki 305-8568, Japan.
| | | | | | | | | |
Collapse
|
34
|
Gilbert ME, Paczkowski C. Propylthiouracil (PTU)-induced hypothyroidism in the developing rat impairs synaptic transmission and plasticity in the dentate gyrus of the adult hippocampus. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2003; 145:19-29. [PMID: 14519490 DOI: 10.1016/s0165-3806(03)00191-3] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Reductions in thyroid hormone during critical periods of brain development can have devastating effects on neurological function that are permanent. Neurochemical, molecular and structural alterations in a variety of brain regions have been well documented, but little information is available on the consequences of developmental hypothyroidism on synaptic function. Developing rats were exposed to the thyrotoxicant, propylthiouracil (PTU: 0 or 15 ppm), through the drinking water of pregnant dams beginning on GD18 and extending throughout the lactational period. Male offspring were allowed to mature after termination of PTU exposure at weaning on PND21 and electrophyiological assessments of field potentials in the dentate gyrus were conducted under urethane anesthesia between 2 and 5 months of age. PTU dramatically reduced thyroid hormones on PND21 and produced deficits in body weight that persisted to adulthood. Synaptic transmission was impaired as evidenced by reductions in excitatory postsynaptic potential (EPSP) slope and population spike (PS) amplitudes at a range of stimulus intensities. Long-term potentiation of the EPSP slope was impaired at both modest and strong intensity trains, whereas a paradoxical increase in PS amplitude was observed in PTU-treated animals in response to high intensity trains. These data are the first to describe functional impairments in synaptic transmission and plasticity in situ as a result of PTU treatment and suggest that perturbations in synaptic function may contribute to learning deficits associated with developmental hypothyroidism.
Collapse
Affiliation(s)
- M E Gilbert
- Neurotoxicology Division (MD-B105-05), National Health and Environmental Effects Laboratory, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | | |
Collapse
|
35
|
Wu J, Huang KP, Huang FL. Participation of NMDA-mediated phosphorylation and oxidation of neurogranin in the regulation of Ca2+- and Ca2+/calmodulin-dependent neuronal signaling in the hippocampus. J Neurochem 2003; 86:1524-33. [PMID: 12950461 DOI: 10.1046/j.1471-4159.2003.01963.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Neurogranin/RC3 (Ng) is a postsynaptic protein kinase C (PKC) substrate and calmodulin (CaM)-binding protein whose CaM-binding affinity is modulated by Ca2+, phosphorylation and oxidation. Ng has been implicated in the modulation of postsynaptic signal transduction pathways and synaptic plasticity. Previously, we showed a severe deficit of spatial memory in Ng knockout (KO) mice. Activation of the NMDA receptor and its downstream signaling molecules are known to be involved in long-term memory formation. In the present study, using mouse hippocampal slices, we demonstrated that NMDA induced a rapid and transient phosphorylation and oxidation of Ng. NMDA also caused activation of PKC as evidenced by their phosphorylations, whereas, such activations were greatly reduced in the KO mice. A higher degree of phosphorylation of Ca2+/CaM-dependent kinase II and activation of cyclic AMP-dependent protein kinase were also evident in the WT compared to those of the KO mice. Phosphorylation of downstream targets, including mitogen-activated protein kinases and cAMP response element-binding protein, were significantly attenuated in the KO mice. These results suggest that by its Ca2+-sensitive CaM-binding feature, and through its phosphorylation and oxidation, Ng regulates the Ca2+- and Ca2+/CaM-dependent signaling pathways subsequent to the stimulation of NMDA receptor. These findings support the hypothesis that the derangement of hippocampal signal transduction cascades in Ng KO mice causes the deficits in synaptic plasticity, learning and memory that occur in these mice.
Collapse
Affiliation(s)
- Junfang Wu
- Section on Metabolic Regulation, Endocrinology and Reproduction Research Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | | | | |
Collapse
|
36
|
van Dalen JJW, Gerendasy DD, de Graan PNE, Schrama LH, Gruol DL. Calcium dynamics are altered in cortical neurons lacking the calmodulin-binding protein RC3. Eur J Neurosci 2003; 18:13-22. [PMID: 12859333 DOI: 10.1046/j.1460-9568.2003.02720.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
RC3 is a neuronal calmodulin-binding protein and protein kinase C substrate that is thought to play an important regulatory role in synaptic transmission and neuronal plasticity. Two molecules known to regulate synaptic transmission and neuronal plasticity are Ca(2+) and calmodulin, and proposed mechanisms of RC3 action involve both molecules. However, physiological evidence for a role of RC3 in neuronal Ca(2+) dynamics is limited. In the current study we utilized cultured cortical neurons obtained from RC3 knockout (RC3-/-) and wildtype mice (RC3+/+) and fura-2-based microscopic Ca(2+) imaging to investigate a role for RC3 in neuronal Ca(2+) dynamics. Immunocytochemical characterization showed that the RC3-/- cultures lack RC3 immunoreactivity, whereas cultures prepared from wildtype mice showed RC3 immunoreactivity at all ages studied. RC3+/+ and RC3-/- cultures were indistinguishable with respect to neuron density, neuronal morphology, the formation of extensive neuritic networks and the presence of glial fibrillary acidic protein (GFAP)-positive astrocytes and gamma-aminobutyric acid (GABA)ergic neurons. However, the absence of RC3 in the RC3-/- neurons was found to alter neuronal Ca(2+) dynamics including baseline Ca(2+) levels measured under normal physiological conditions or after blockade of synaptic transmission, spontaneous intracellular Ca(2+) oscillations generated by network synaptic activity, and Ca(2+) responses elicited by exogenous application of N-methyl-D-aspartate (NMDA) or class I metabotropic glutamate receptor agonists. Thus, significant changes in Ca(2+) dynamics occur in cortical neurons when RC3 is absent and these changes do not involve changes in gross neuronal morphology or neuronal maturation. These data provide direct physiological evidence for a regulatory role of RC3 in neuronal Ca(2+) dynamics.
Collapse
Affiliation(s)
- Jacqueline J W van Dalen
- Division of Pharmacology and Anatomy, Rudolf Magnus Institute for Neurosciences, University Medical Center, Utrecht, The Netherlands
| | | | | | | | | |
Collapse
|
37
|
Goldin M, Segal M. Protein kinase C and ERK involvement in dendritic spine plasticity in cultured rodent hippocampal neurons. Eur J Neurosci 2003; 17:2529-39. [PMID: 12823460 DOI: 10.1046/j.1460-9568.2003.02694.x] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The roles of protein kinase C and the MAP-kinase extracellular receptor kinase in structural changes associated with long-term potentiation of network activity were examined in cultured hippocampal neurons. A brief exposure to a conditioning medium that favours activation of the N-methyl-d-aspartate receptor caused a rapid and specific increase in staining of neurons for the phosphorylated form of extracellular receptor kinase as well as of cyclic AMP response element binding protein. Exposure of the cultures to the conditioning medium was followed by a protein synthesis-dependent formation of novel dendritic spines. An extracellular receptor kinase antagonist PD98059 blocked the phosphorylated form of extracellular receptor kinase response and the formation of novel spines. A selective protein kinase C agonist, phorbol 12-myristate 13-acetate, caused activation of extracellular receptor kinase and formation of novel spines. The protein kinase C antagonist GF109203x blocked the phosphorylated form of extracellular receptor kinase response and the subsequent spine formation by phorbol 12-myristate 13-acetate. Both the conditioning medium and phorbol 12-myristate 13-acetate caused a delayed increase in mean amplitude of miniature excitatory postsynaptic currents recorded in the hippocampal neurons. These results indicate that activation of extracellular receptor kinase mediates the effect of a conditioning protocol on the formation of dendritic spines. The formation of novel spines was associated with long-term increase in network activity and functional synaptic connectivity among the cultured neurons.
Collapse
Affiliation(s)
- Miri Goldin
- Department of Neurobiology, The Weizmann Institute, Rehovot 76100, Israel
| | | |
Collapse
|
38
|
Singec I, Knoth R, Ditter M, Frotscher M, Volk B. Neurogranin expression by cerebellar neurons in rodents and non-human primates. J Comp Neurol 2003; 459:278-89. [PMID: 12655510 DOI: 10.1002/cne.10600] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Neurogranin (NG) is a brain-specific protein kinase C substrate involved in the regulation of calcium signaling and neuronal plasticity. A rostrocaudal expression profile, with large amounts in telencephalic brain regions and low expression levels in phylogenetically older brain structures, was reported previously. In the cerebellum, expression of NG has not been described. By using immunocytochemistry and in situ hybridization, we found that NG is expressed in the mouse (C57Bl/6), rat (Wistar), and monkey (Cercopithecus aetiops) cerebella. In the mouse cerebellum, Golgi cells were strongly immunoreactive for NG, whereas other cerebellar neurons were devoid of this protein. Cell counts showed 1.6-fold more immunopositive Golgi cells in the hemispheres (61.1 +/- 8.0 cells/mm(2)) than in the vermis (37.5 +/- 3.3 cells/mm(2)). Developmental studies showed detectable NG in the mouse cerebellum as early as on postnatal day 10 (P10). In contrast to the mouse, in the rat cerebellum we found only a few Golgi cells containing NG (hemispheres, 2.4 +/- 0.5 cells/mm(2); vermis, 1.5 +/- 0.3 cells/mm(2)). In the monkey cerebellum, unipolar brush cells, localized in the granular layer, were heavily labeled, whereas Golgi cells were devoid of NG. This study demonstrated that NG is strongly expressed in specific gamma-aminobutyric acidergic neurons in the rodent cerebellum. In addition, NG expression in the primate cerebellum by brush cells, which are excitatory, showed remarkable cell type-specific and species-specific expression patterns of a postsynaptic protein mediating calcium signaling mechanisms.
Collapse
Affiliation(s)
- Ilyas Singec
- Institute of Anatomy, University of Freiburg, D-79104 Freiburg, Germany.
| | | | | | | | | |
Collapse
|
39
|
Yang JH, Derr-Yellin EC, Kodavanti PRS. Alterations in brain protein kinase C isoforms following developmental exposure to a polychlorinated biphenyl mixture. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2003; 111:123-35. [PMID: 12654512 DOI: 10.1016/s0169-328x(02)00697-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
PCBs have been shown to alter several neurochemical end-points and are implicated in the etiology of some neurological diseases. Recent in vivo studies from our laboratory indicated that developmental exposure to a commercial PCB mixture, Aroclor 1254, caused perturbations in calcium homeostasis and changes in protein kinase C (PKC) activities in rat brain. However, it is not known which molecular substances are targets for PCB-induced developmental neurotoxicity. Since the PKC signaling pathway has been implicated in the modulation of motor behavior as well as learning and memory, and the roles of PKC are subspecies specific, the present study attempted to analyze the effects on selected PKC isozymes in the cerebellum and the hippocampus following developmental exposure (gestational day 6 through postnatal day 21) to a PCB mixture, Aroclor 1254. The results indicated that the developmental exposure to PCBs caused significant hypothyroxinemia and age-dependent alterations in the translocation of PKC isozymes; the effects were greatly significant at postnatal day (PND) 14. Immunoblot analysis of PKC-alpha (alpha) from both cerebellum and hippocampus revealed that developmental exposure to Aroclor 1254 caused a significant decrease in cytosolic fraction and an increase in particulate fraction. There was no significant difference between these two brain regions on the level of fractional changes. However, the ratio between the fractions (particulate/cytosol) from cerebellum only was increased in a dose-dependent manner. Analysis of PKC-gamma (gamma) in cerebellum on PND14 showed a decrease in cytosolic fraction in both dose groups and an increase in particulate fraction at high dose (6 mg/kg) only. The ratio between the two fractions was increased in a dose-dependent manner. In the hippocampus, there was a significant decrease in PKC-gamma in cytosolic fraction of the high-dose group and a significant increase in particulate fraction of the low-dose group. But, the ratio between the fractions showed a significant increase (2.6-fold increase in high dose on PND14). Analysis of PKC-epsilon (epsilon) in cerebellum showed a significant decrease in cytosolic fraction at PND14, while particulate PKand an increase in ratio between fractions at 6 mg/kg on PND14. The results from this study indicate that the patterns of subcellular distributions of PKC isoforms following a developmental PCB exposure were PKC isozyme- and developmental stage-specific. Considering the significant role of PKC signaling in motor behavior, learning and memory, it is suggested that altered subcellular distribution of PKC isoforms at critical periods of brain development may be a possible mechanism of PCB-induced neurotoxic effects and that PKC-alpha, gamma, and epsilon may be among the target molecules implicated with PCB-induced neurological impairments during developmental exposure. It is believed that this is the first report successfully identifying PKC isoforms responding to PCBs during developmental exposure.
Collapse
Affiliation(s)
- Jae-Ho Yang
- National Academy of Sciences, National Research Council, Washington, DC, USA
| | | | | |
Collapse
|
40
|
Foster TC. Regulation of synaptic plasticity in memory and memory decline with aging. PROGRESS IN BRAIN RESEARCH 2002; 138:283-303. [PMID: 12432775 DOI: 10.1016/s0079-6123(02)38083-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Thomas C Foster
- Department of Molecular and Biomedical Pharmacology, University of Kentucky, College of Medicine, Lexington, KY 40536, USA.
| |
Collapse
|
41
|
Abstract
Mood stabilizers represent a class of drugs that are efficacious in the treatment of bipolar disorder. The most established medications in this class are lithium, valproic acid, and carbamazepine. In addition to their therapeutic effects for treatment of acute manic episodes, these medications often are useful as prophylaxis against future episodes and as adjunctive antidepressant medications. While important extracellular effects have not been excluded, most available evidence suggests that the therapeutically relevant targets of this class of medications are in the interior of cells. Herein we give a prospective of a rapidly evolving field, discussing common effects of mood stabilizers as well as effects that are unique to individual medications. Mood stabilizers have been shown to modulate the activity of enzymes, ion channels, arachidonic acid turnover, G protein coupled receptors and intracellular pathways involved in synaptic plasticity and neuroprotection. Understanding the therapeutic targets of mood stabilizers will undoubtedly lead to a better understanding of the pathophysiology of bipolar disorder and to the development of improved therapeutics for the treatment of this disease. Furthermore, the involvement of mood stabilizers in pathways operative in neuroprotection suggests that they may have utility in the treatment of classical neurodegenerative disorders.
Collapse
Affiliation(s)
- Todd D. Gould
- Laboratory of Molecular Pathophysiology, Building 49, Room B1EE16, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Guang Chen
- Laboratory of Molecular Pathophysiology, Building 49, Room B1EE16, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Husseini K. Manji
- Laboratory of Molecular Pathophysiology, Building 49, Room B1EE16, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
42
|
Abstract
In mammals, long-term memory induced by Pavlovian fear conditioning has been shown to be dependent on the amygdala during a protein and mRNA synthesis-dependent phase of memory consolidation. We have used genes identified in a kainic acid model of synaptic plasticity as in situ hybridization probes during the consolidation period after fear conditioning. We found that these genes were transcriptionally regulated in several brain areas only when stimuli were presented in a manner that supported behavioral learning and not after unpaired presentations or footshocks alone. Immediate early genes and neurofilament mRNA peaked approximately 30 min after conditioning, as expected. Interestingly, nurr-1, alpha-actinin, and 16c8 increased approximately 2-4 hr later, whereas neurogranin and gephyrin decreased during that time. Our results suggest that fear memory consolidation occurs within a broad neural circuit that includes, but is not limited to, the amygdala. Together, a broad array of transcriptionally regulated genes, encoding transcription factors, cytoskeletal proteins, adhesion molecules, and receptor stabilization molecules, appear to mediate the neural plasticity underlying specific forms of long-term memory in mammals.
Collapse
|
43
|
Miyakawa T, Yared E, Pak JH, Huang FL, Huang KP, Crawley JN. Neurogranin null mutant mice display performance deficits on spatial learning tasks with anxiety related components. Hippocampus 2002; 11:763-75. [PMID: 11811671 DOI: 10.1002/hipo.1092] [Citation(s) in RCA: 144] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Neurogranin/RC3 is a protein that binds calmodulin and serves as a substrate for protein kinase C. Neuronally distributed in the hippocampus and forebrain, neurogranin is highly expressed in dendritic spines of hippocampal pyramidal cells, implicating this protein in long-term potentiation and in learning and memory processes. Null mutation of the neurogranin gene Ng generated viable knockout mice for analysis of the behavioral phenotype resulting from the absence of neurogranin protein. Ng -/- mice were normal on measures of general health, neurological reflexes, sensory abilities, and motor functions, as compared to wild type littermate controls. On the Morris water task, Ng -/- mice failed to reach acquisition criterion on the hidden platform test and did not show selective search on the probe trial. In the Barnes circular maze, another test for spatial navigation learning, Ng -/- mice showed impairments on some components of transfer, but normal performance on time spent around the target hole. Abnormal and idiosyncratic behaviors were detected, that appeared to represent an anxiogenic phenotype in Ng -/- mice, as measured in the light<-->dark exploration test and the open field center time parameter. These findings of apparent deficits in spatial learning and anxiety-like tendencies in Ng -/- support a role for neurogranin in the hippocampally-mediated interaction between stress and performance.
Collapse
Affiliation(s)
- T Miyakawa
- Section on Behavioral Neuropharmacology, Experimental Therapeutics Branch, National Institute of Mental Health, Bethesda, Maryland 20892-1375, USA
| | | | | | | | | | | |
Collapse
|
44
|
Mons N, Enderlin V, Jaffard R, Higueret P. Selective age-related changes in the PKC-sensitive, calmodulin-binding protein, neurogranin, in the mouse brain. J Neurochem 2001; 79:859-67. [PMID: 11723178 DOI: 10.1046/j.1471-4159.2001.00646.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Brain ageing is associated with a dysregulation of intracellular calcium (Ca(2+)) homeostasis which leads to deficits in Ca(2+)-dependent signalling pathways and altered neuronal functions. Given the crucial role of neurogranin/RC3 (Ng) in the post-synaptic regulation of Ca(2+) and calmodulin levels, age-dependent changes in the levels of Ng mRNA and protein expression were analysed in 3, 12, 24 and 31-month-old mouse brains. Ageing produced significant decreases in Ng mRNA expression in the dorsal hippocampal subfields, retrosplenial and primary motor cortices, whereas no reliable changes were seen in any other cortical regions examined. Western blot indicated that Ng protein expression was also down-regulated in the ageing mouse brain. Analysis of Ng immunoreactivity in both hippocampal CA1 and retrosplenial areas indicated that Ng protein in aged mice decreased predominantly in the dendritic segments of pyramidal neurones. These data suggest that age-related changes of post-synaptic Ng in selected brain areas, and particularly in hippocampus, may contribute to altered Ca(2+)/calmodulin-signalling pathways and to region-specific impairments of synaptic plasticity and cognitive decline.
Collapse
Affiliation(s)
- N Mons
- Laboratoire de Neurosciences Cognitives UMR CNRS 5106, Université de Bordeaux, Talence, France.
| | | | | | | |
Collapse
|
45
|
Valjent E, Caboche J, Vanhoutte P. Mitogen-activated protein kinase/extracellular signal-regulated kinase induced gene regulation in brain: a molecular substrate for learning and memory? Mol Neurobiol 2001; 23:83-99. [PMID: 11817219 DOI: 10.1385/mn:23:2-3:083] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) pathway is an evolutionarily conserved signaling cascade involved in a plethora of physiological responses, including cell proliferation, survival, differentiation, and, in neuronal cells, synaptic plasticity. Increasing evidence now implicates this pathway in cognitive functions, such as learning and memory formation, and also in behavioral responses to addictive drugs. Although multiple intracellular substrates can be activated by ERKs, nuclear targeting of transcription factors, and thereby control of gene expression, seems to be a major event in ERK-induced neuronal adaptation. By controlling a prime burst of gene expression, ERK signaling could be critically involved in molecular adaptations that are necessary for long-term behavioral changes. Reviewed here are data providing evidence for a role of ERKs in long-term behavioral alterations, and the authors discuss molecular mechanisms that could underlie this role.
Collapse
Affiliation(s)
- E Valjent
- Laboratoire de Signalisation Neuronale et Régulations Géniques, CNRS-Université Pierre et Marie Curie, Paris, France
| | | | | |
Collapse
|
46
|
Bennett PC, Zhao W, Ng KT. Concentration-dependent effects of protein phosphatase (PP) inhibitors implicate PP1 and PP2A in different stages of memory formation. Neurobiol Learn Mem 2001; 75:91-110. [PMID: 11124049 DOI: 10.1006/nlme.1999.3959] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Numerous studies have demonstrated roles for protein phosphorylation and for specific kinases in memory formation; however, a role for specific protein phosphatases has not been established. Previous studies using pharmacobehavioral methods to implicate protein phosphatase activity in memory formation have been unable to discriminate between protein phosphatases 1 (PP1) and 2A (PP2A), as available cell-permeable agents generally inhibit both enzyme classes. To address this difficulty the present study exploited differences in the potency of the selective phosphatase inhibitor, okadaic acid, toward PP1 and PP2A. Within the context of a temporally precise animal model of memory, developed using the day-old chick (Gallus domesticus), acute administration of various concentrations of okadaic acid was found to disrupt two temporally distinct stages of memory formation. When administered bilaterally into an area of the chick brain implicated in memory formation, concentrations of okadaic acid known to selectively inhibit PP2A in vitro disrupted memory from 50 min posttraining. Higher concentrations, reported to inhibit both PP2A and PP1 in vitro, produced significant retention deficits from 20 min posttraining. Identical temporally specific effects were also obtained by varying the concentration and time of administration of calyculin A, a phosphatase inhibitor with equal potency toward both enzyme classes. Hence, different phosphatase enzymes may contribute to different stages of the enzymatic cascade believed to underlie memory formation.
Collapse
Affiliation(s)
- P C Bennett
- Department of Psychology, Monash University, Clayton, Victoria, 3168, Australia
| | | | | |
Collapse
|
47
|
Rampon C, Jiang CH, Dong H, Tang YP, Lockhart DJ, Schultz PG, Tsien JZ, Hu Y. Effects of environmental enrichment on gene expression in the brain. Proc Natl Acad Sci U S A 2000; 97:12880-4. [PMID: 11070096 PMCID: PMC18858 DOI: 10.1073/pnas.97.23.12880] [Citation(s) in RCA: 450] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
An enriched environment is known to promote structural changes in the brain and to enhance learning and memory performance in rodents [Hebb, D. O. (1947) Am. Psychol. 2, 306-307]. To better understand the molecular mechanisms underlying these experience-dependent cognitive changes, we have used high-density oligonucleotide microarrays to analyze gene expression in the brain. Expression of a large number of genes changes in response to enrichment training, many of which can be linked to neuronal structure, synaptic plasticity, and transmission. A number of these genes may play important roles in modulating learning and memory capacity.
Collapse
Affiliation(s)
- C Rampon
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121; and the Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Dowling AL, Zoeller RT. Thyroid hormone of maternal origin regulates the expression of RC3/neurogranin mRNA in the fetal rat brain. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2000; 82:126-32. [PMID: 11042365 DOI: 10.1016/s0169-328x(00)00190-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Recent clinical studies indicate that thyroid hormone plays essential roles in fetal brain development. However, the mechanism by which thyroid hormone affects fetal brain development is poorly studied. We recently identified several genes expressed in the fetal cortex whose abundance is affected by thyroid hormone of maternal origin. However, it is unclear whether these genes are directly regulated by thyroid hormone. Because these are the first genes known to be regulated by thyroid hormone during fetal development, we sought to expand our investigation to genes known to be regulated directly by thyroid hormone. We now report that the well-known thyroid hormone-responsive gene RC3/neurogranin is expressed in the fetal brain and is regulated by thyroid hormone of maternal origin. These findings support the concept that maternal thyroid hormone exerts a direct action on the expression of genes in the fetal brain that are important for normal neurological development.
Collapse
Affiliation(s)
- A L Dowling
- Biology Department and Molecular and Cellular Biology Program, Morrill Science Center, University of Massachusetts, Amherst, MA 01003, USA
| | | |
Collapse
|
49
|
Abstract
The protein kinase C family of enzymes has been implicated in synaptic plasticity and memory in a wide range of animal species, but to date little information has been available concerning specific roles for individual isoforms of this category of kinases. To investigate the role of the beta isoform of PKC in mammalian learning, we characterized mice deficient in the PKC beta gene using anatomical, biochemical, physiological, and behavioral approaches. In our studies we observed that PKC beta was predominantly expressed in the neocortex, in area CA1 of the hippocampus, and in the basolateral nucleus of the amygdala. Mice deficient in PKC beta showed normal brain anatomy and normal hippocampal synaptic transmission, paired pulse facilitation, and long-term potentiation and normal sensory and motor responses. The PKC beta knock-out animals exhibited a loss of learning, however; they suffered deficits in both cued and contextual fear conditioning. The PKC expression pattern and behavioral phenotype in the PKC beta knock-out animals indicate a critical role for the beta isoform of PKC in learning-related signal transduction mechanisms, potentially in the basolateral nucleus of the amygdala.
Collapse
|
50
|
Muñoz MD, Monfort P, Gaztelu JM, Felipo V. Hyperammonemia impairs NMDA receptor-dependent long-term potentiation in the CA1 of rat hippocampus in vitro. Neurochem Res 2000; 25:437-41. [PMID: 10823575 DOI: 10.1023/a:1007547622844] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Hyperammonemia is considered the main factor responsible for the neurological and cognitive alterations found in hepatic encephalopathy and in patients with congenital deficiencies of the urea cycle enzymes. The underlying mechanisms remain unclear. Chronic moderate hyperammonemia reduces nitric oxide-induced activation of soluble guanylate cyclase and glutamate-induced formation of cGMP. NMDA receptor-associated transduction pathways, including activation of soluble guanylate cyclase, are involved in the induction of long-term potentiation (LTP), a phenomenon that is considered to be the molecular basis for some forms of memory and learning. Using an animal model we show that chronic hyperammonemia significantly reduces the degree of long-term potentiation induced in the CA1 of hippocampus slices (200% increase in control and 50% increase in slices of hyperammonemic animals). Also, addition of 1 mM ammonia impaired the maintenance of non-decremental LTP. The LTP impairment could be involved in the intellectual impairment present in chronic hepatocerebral disorders associated with hyperammonemia.
Collapse
Affiliation(s)
- M D Muñoz
- Neurología Experimental (Unidad Asociada al CSIC), Departamento de Investigación, Hospital Ramón y Cajal, Madrid, Spain.
| | | | | | | |
Collapse
|