1
|
Richter N, David LS, Grothe MJ, Teipel S, Dietlein M, Tittgemeyer M, Neumaier B, Fink GR, Onur OA, Kukolja J. Age and Anterior Basal Forebrain Volume Predict the Cholinergic Deficit in Patients with Mild Cognitive Impairment due to Alzheimer’s Disease. J Alzheimers Dis 2022; 86:425-440. [DOI: 10.3233/jad-210261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Background: Early and severe neuronal loss in the cholinergic basal forebrain is observed in Alzheimer’s disease (AD). To date, cholinomimetics play a central role in the symptomatic treatment of AD dementia. Although basic research indicates that a cholinergic deficit is present in AD before dementia, the efficacy of cholinomimetics in mild cognitive impairment (MCI) remains controversial. Predictors of cholinergic impairment could guide individualized therapy. Objective: To investigate if the extent of the cholinergic deficit, measured using positron emission tomography (PET) and the tracer 11C-N-methyl-4-piperidyl acetate (MP4A), could be predicted from the volume of cholinergic basal forebrain nuclei in non-demented AD patients. Methods: Seventeen patients with a high likelihood of MCI due to AD and 18 age-matched cognitively healthy adults underwent MRI-scanning. Basal forebrain volume was assessed using voxel-based morphometry and a cytoarchitectonic atlas of cholinergic nuclei. Cortical acetylcholinesterase (AChE) activity was measured using MP4A-PET. Results: Cortical AChE activity and nucleus basalis of Meynert (Ch4 area) volume were significantly decreased in MCI. The extent of the cholinergic deficit varied considerably across patients. Greater volumes of anterior basal forebrain nuclei (Ch1/2 area) and younger age (Spearman’s rho (17) = –0.596, 95% -CI [–0.905, –0.119] and 0.593, 95% -CI [0.092, 0.863])) were associated with a greater cholinergic deficit. Conclusion: Data suggest that less atrophy of the Ch1/2 area and younger age are associated with a more significant cholinergic deficit in MCI due to AD. Further investigations are warranted to determine if the individual response to cholinomimetics can be inferred from these measures.
Collapse
Affiliation(s)
- Nils Richter
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, Jülich, Germany
- Department of Neurology, Medical Faculty and University Hospital of Cologne, Cologne, Germany
- Max-Planck-Institute for Metabolism Research, Cologne, Cologne, Germany
| | - Lara-Sophia David
- Department of Neurology, Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - Michel J. Grothe
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Movement Disorders Group, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| | - Markus Dietlein
- Department of Nuclear Medicine, Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - Marc Tittgemeyer
- Max-Planck-Institute for Metabolism Research, Cologne, Cologne, Germany
| | - Bernd Neumaier
- Max-Planck-Institute for Metabolism Research, Cologne, Cologne, Germany
- Nuclear Chemistry, Institute of Neuroscience and Medicine (INM-5), Research Center Jülich, Jülich, Germany
- Institute for Radiochemistry and Experimental Molecular Imaging, Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - Gereon R. Fink
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, Jülich, Germany
- Department of Neurology, Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - Oezguer A. Onur
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, Jülich, Germany
- Department of Neurology, Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - Juraj Kukolja
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, Jülich, Germany
- Department of Neurology, Medical Faculty and University Hospital of Cologne, Cologne, Germany
- Department of Neurology and Clinical Neurophysiology, Helios University Hospital Wuppertal, Wuppertal, Germany
- Faculty of Health, Witten/Herdecke University, Witten, Germany
| |
Collapse
|
2
|
Roggenhofer E, Toumpouli E, Seeck M, Wiest R, Lutti A, Kherif F, Novy J, Rossetti AO, Draganski B. Clinical phenotype modulates brain's myelin and iron content in temporal lobe epilepsy. Brain Struct Funct 2021; 227:901-911. [PMID: 34817680 PMCID: PMC8930791 DOI: 10.1007/s00429-021-02428-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 11/09/2021] [Indexed: 11/17/2022]
Abstract
Temporal lobe epilepsy (TLE) is associated with brain pathology extending beyond temporal lobe structures. We sought to look for informative patterns of brain tissue properties in TLE that go beyond the established morphometry differences. We hypothesised that volume differences, particularly in hippocampus, will be paralleled by changes in brain microstructure. The cross-sectional study included TLE patients (n = 25) from a primary care center and sex-/age-matched healthy controls (n = 55). We acquired quantitative relaxometry-based magnetic resonance imaging (MRI) data yielding whole-brain maps of grey matter volume, magnetization transfer (MT) saturation, and effective transverse relaxation rate R2* indicative for brain tissue myelin and iron content. For statistical analysis, we used the computational anatomy framework of voxel-based morphometry and voxel-based quantification. There was a positive correlation between seizure activity and MT saturation measures in the ipsilateral hippocampus, paralleled by volume differences bilaterally. Disease duration correlated positively with iron content in the mesial temporal lobe, while seizure freedom was associated with a decrease of iron in the very same region. Our findings demonstrate the link between TLE clinical phenotype and brain anatomy beyond morphometry differences to show the impact of disease burden on specific tissue properties. We provide direct evidence for the differential effect of clinical phenotype characteristics on processes involving tissue myelin and iron in mesial temporal lobe structures. This study offers a proof-of-concept for the investigation of novel imaging biomarkers in focal epilepsy.
Collapse
Affiliation(s)
- Elisabeth Roggenhofer
- LREN, Centre for Research in Neuroscience, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Mont Paisible 16, 1011, Lausanne, Switzerland.,EEG and Epilepsy Unit, Department of Neurology, Department of Clinical Neurosciences, University Hospitals and Faculty of Medicine Geneva, Geneva, Switzerland
| | - Evdokia Toumpouli
- LREN, Centre for Research in Neuroscience, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Mont Paisible 16, 1011, Lausanne, Switzerland
| | - Margitta Seeck
- EEG and Epilepsy Unit, Department of Neurology, Department of Clinical Neurosciences, University Hospitals and Faculty of Medicine Geneva, Geneva, Switzerland
| | - Roland Wiest
- Support Center for Advanced Neuroimaging, Institute for Diagnostic and Interventional Neuroradiology, University Hospital Inselspital, University of Bern, Bern, Switzerland
| | - Antoine Lutti
- LREN, Centre for Research in Neuroscience, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Mont Paisible 16, 1011, Lausanne, Switzerland
| | - Ferath Kherif
- LREN, Centre for Research in Neuroscience, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Mont Paisible 16, 1011, Lausanne, Switzerland
| | - Jan Novy
- Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Andrea O Rossetti
- Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Bogdan Draganski
- LREN, Centre for Research in Neuroscience, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Mont Paisible 16, 1011, Lausanne, Switzerland. .,Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland. .,Department of Neurology, Max-Planck-Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
| |
Collapse
|
3
|
Richter N, Beckers N, Onur OA, Dietlein M, Tittgemeyer M, Kracht L, Neumaier B, Fink GR, Kukolja J. Effect of cholinergic treatment depends on cholinergic integrity in early Alzheimer's disease. Brain 2019; 141:903-915. [PMID: 29309600 DOI: 10.1093/brain/awx356] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/08/2017] [Indexed: 02/04/2023] Open
Abstract
In early Alzheimer's disease, which initially presents with progressive loss of short-term memory, neurodegeneration especially affects cholinergic neurons of the basal forebrain. Pharmacotherapy of Alzheimer's disease therefore often targets the cholinergic system. In contrast, cholinergic pharmacotherapy of mild cognitive impairment is debated since its efficacy to date remains controversial. We here investigated the relationship between cholinergic treatment effects and the integrity of the cholinergic system in mild cognitive impairment due to Alzheimer's disease. Fourteen patients with high likelihood of mild cognitive impairment due to Alzheimer's disease and 16 age-matched cognitively normal adults performed an episodic memory task during functional magnetic resonance imaging under three conditions: (i) without pharmacotherapy; (ii) with placebo; and (iii) with a single dose of rivastigmine (3 mg). Cortical acetylcholinesterase activity was measured using PET with the tracer 11C-N-methyl-4-piperidyl acetate (MP4A). Cortical acetylcholinesterase activity was significantly decreased in patients relative to controls, especially in the lateral temporal lobes. Without pharmacotherapy, mild cognitive impairment was associated with less memory-related neural activation in the fusiform gyrus and impaired deactivation in the posterior cingulate cortex, relative to controls. These differences were attenuated under cholinergic stimulation with rivastigmine: patients showed increased neural activation in the right fusiform gyrus but enhanced deactivation of the posterior cingulate cortex under rivastigmine, compared to placebo. Conversely, controls showed reduced activation of the fusiform gyrus and reduced deactivation of the posterior cingulate under rivastigmine, compared to placebo. In both groups, the change in neural activation in response to rivastigmine was negatively associated with local acetylcholinesterase activity. At the behavioural level, an analysis of covariance revealed a significant group × treatment interaction in episodic memory performance when accounting for hippocampal grey matter atrophy and function. Our results indicate that rivastigmine differentially affects memory-related neural activity in patients with mild cognitive impairment and cognitively normal, age-matched adults, depending on acetylcholinesterase activity as a marker for the integrity of the cortical cholinergic system. Furthermore, hippocampal integrity showed an independent association with the response of memory performance to acetylcholinesterase inhibition.
Collapse
Affiliation(s)
- Nils Richter
- Department of Neurology, University Hospital Cologne, 50937 Cologne, Germany.,Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany.,Max-Planck-Institute for Metabolism Research, 50937 Cologne, Germany
| | - Nora Beckers
- Department of Neurology, University Hospital Cologne, 50937 Cologne, Germany
| | - Oezguer A Onur
- Department of Neurology, University Hospital Cologne, 50937 Cologne, Germany.,Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany
| | - Markus Dietlein
- Department of Nuclear Medicine, University Hospital Cologne, 50937 Cologne, Germany
| | - Marc Tittgemeyer
- Max-Planck-Institute for Metabolism Research, 50937 Cologne, Germany
| | - Lutz Kracht
- Max-Planck-Institute for Metabolism Research, 50937 Cologne, Germany.,Department of Nuclear Medicine, University Hospital Cologne, 50937 Cologne, Germany
| | - Bernd Neumaier
- Nuclear Chemistry, Institute of Neuroscience and Medicine (INM-5), Research Center Jülich, 52425 Jülich, Germany.,Institute for Radiochemistry and Experimental Molecular Imaging, University Hospital Cologne, 50937 Cologne, Germany
| | - Gereon R Fink
- Department of Neurology, University Hospital Cologne, 50937 Cologne, Germany.,Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany
| | - Juraj Kukolja
- Department of Neurology, University Hospital Cologne, 50937 Cologne, Germany.,Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany
| |
Collapse
|
4
|
Gil V, Del Río JA. Functions of Plexins/Neuropilins and Their Ligands during Hippocampal Development and Neurodegeneration. Cells 2019; 8:E206. [PMID: 30823454 PMCID: PMC6468495 DOI: 10.3390/cells8030206] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 02/22/2019] [Accepted: 02/24/2019] [Indexed: 12/22/2022] Open
Abstract
There is emerging evidence that molecules, receptors, and signaling mechanisms involved in vascular development also play crucial roles during the development of the nervous system. Among others, specific semaphorins and their receptors (neuropilins and plexins) have, in recent years, attracted the attention of researchers due to their pleiotropy of functions. Their functions, mainly associated with control of the cellular cytoskeleton, include control of cell migration, cell morphology, and synapse remodeling. Here, we will focus on their roles in the hippocampal formation that plays a crucial role in memory and learning as it is a prime target during neurodegeneration.
Collapse
Affiliation(s)
- Vanessa Gil
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Parc Científic de Barcelona, 08028 Barcelona, Spain.
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, 08028 Barcelona, Spain.
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), 08028 Barcelona, Spain.
- Institute of Neuroscience, University of Barcelona, 08028 Barcelona, Spain.
| | - José Antonio Del Río
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Parc Científic de Barcelona, 08028 Barcelona, Spain.
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, 08028 Barcelona, Spain.
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), 08028 Barcelona, Spain.
- Institute of Neuroscience, University of Barcelona, 08028 Barcelona, Spain.
| |
Collapse
|
5
|
Mata A, Gil V, Pérez-Clausell J, Dasilva M, González-Calixto MC, Soriano E, García-Verdugo JM, Sanchez-Vives MV, Del Río JA. New functions of Semaphorin 3E and its receptor PlexinD1 during developing and adult hippocampal formation. Sci Rep 2018; 8:1381. [PMID: 29358640 PMCID: PMC5777998 DOI: 10.1038/s41598-018-19794-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 01/08/2018] [Indexed: 12/19/2022] Open
Abstract
The development and maturation of cortical circuits relies on the coordinated actions of long and short range axonal guidance cues. In this regard, the class 3 semaphorins and their receptors have been seen to be involved in the development and maturation of the hippocampal connections. However, although the role of most of their family members have been described, very few data about the participation of Semaphorin 3E (Sema3E) and its receptor PlexinD1 during the development and maturation of the entorhino-hippocampal (EH) connection are available. In the present study, we focused on determining their roles both during development and in adulthood. We determined a relevant role for Sema3E/PlexinD1 in the layer-specific development of the EH connection. Indeed, mice lacking Sema3E/PlexinD1 signalling showed aberrant layering of entorhinal axons in the hippocampus during embryonic and perinatal stages. In addition, absence of Sema3E/PlexinD1 signalling results in further changes in postnatal and adult hippocampal formation, such as numerous misrouted ectopic mossy fibers. More relevantly, we describe how subgranular cells express PlexinD1 and how the absence of Sema3E induces a dysregulation of the proliferation of dentate gyrus progenitors leading to the presence of ectopic cells in the molecular layer. Lastly, Sema3E mutant mice displayed increased network excitability both in the dentate gyrus and the hippocampus proper.
Collapse
Affiliation(s)
- Agata Mata
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Parc Científic de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, (CIBERNED), Barcelona, Spain.,Institut de Neurociències de la Universitat de Barcelona, Barcelona, Spain
| | - Vanessa Gil
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Parc Científic de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, (CIBERNED), Barcelona, Spain.,Institut de Neurociències de la Universitat de Barcelona, Barcelona, Spain
| | - Jeús Pérez-Clausell
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain
| | - Miguel Dasilva
- Systems Neuroscience, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Mari Carmen González-Calixto
- Laboratory of Comparative Neurobiology, Institute Cavanilles, University of Valencia, CIBERNED, 46980, Valencia, Spain
| | - Eduardo Soriano
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, (CIBERNED), Barcelona, Spain.,Institut de Neurociències de la Universitat de Barcelona, Barcelona, Spain.,ICREA, Barcelona, Spain.,Vall d'Hebrón Institut de Recerca (VHIR), Barcelona, Spain
| | - José Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Institute Cavanilles, University of Valencia, CIBERNED, 46980, Valencia, Spain
| | - Maria V Sanchez-Vives
- Systems Neuroscience, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,ICREA, Barcelona, Spain
| | - José Antonio Del Río
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Parc Científic de Barcelona, Barcelona, Spain. .,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, (CIBERNED), Barcelona, Spain. .,Institut de Neurociències de la Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
6
|
Plasticity Related Gene 3 (PRG3) overcomes myelin-associated growth inhibition and promotes functional recovery after spinal cord injury. Aging (Albany NY) 2017; 8:2463-2487. [PMID: 27744421 PMCID: PMC5115901 DOI: 10.18632/aging.101066] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/25/2016] [Indexed: 12/14/2022]
Abstract
The Plasticity Related Gene family covers five, brain-specific, transmembrane proteins (PRG1-5, also termed LPPR1-5) that operate in neuronal plasticity during development, aging and brain trauma. Here we investigated the role of the PRG family on axonal and filopodia outgrowth. Comparative analysis revealed the strongest outgrowth induced by PRG3 (LPPR1). During development, PRG3 is ubiquitously located at the tip of neuronal processes and at the plasma membrane and declines with age. In utero electroporation of PRG3 induced dendritic protrusions and accelerated spine formations in cortical pyramidal neurons. The neurite growth promoting activity of PRG3 requires RasGRF1 (RasGEF1/Cdc25) mediated downstream signaling. Moreover, in axon collapse assays, PRG3-induced neurites resisted growth inhibitors such as myelin, Nogo-A (Reticulon/RTN-4), thrombin and LPA and impeded the RhoA-Rock-PIP5K induced neurite repulsion. Transgenic adult mice with constitutive PRG3 expression displayed strong axonal sprouting distal to a spinal cord lesion. Moreover, fostered PRG3 expression promoted complex motor-behavioral recovery compared to wild type controls as revealed in the Schnell swim test (SST). Thus, PRG3 emerges as a developmental RasGRF1-dependent conductor of filopodia formation and axonal growth enhancer. PRG3-induced neurites resist brain injury-associated outgrowth inhibitors and contribute to functional recovery after spinal cord lesions. Here, we provide evidence that PRG3 operates as an essential neuronal growth promoter in the nervous system. Maintaining PRG3 expression in aging brain may turn back the developmental clock for neuronal regeneration and plasticity.
Collapse
|
7
|
The cholinergic system in aging and neuronal degeneration. Behav Brain Res 2011; 221:555-63. [DOI: 10.1016/j.bbr.2010.11.058] [Citation(s) in RCA: 752] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2010] [Accepted: 11/26/2010] [Indexed: 11/19/2022]
|
8
|
Ginsberg SD. Alterations in discrete glutamate receptor subunits in adult mouse dentate gyrus granule cells following perforant path transection. Anal Bioanal Chem 2010; 397:3349-58. [PMID: 20577723 PMCID: PMC3149099 DOI: 10.1007/s00216-010-3826-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Revised: 04/27/2010] [Accepted: 05/04/2010] [Indexed: 01/25/2023]
Abstract
Custom-designed microarray analysis was utilized to evaluate expression levels of glutamate receptors (GluRs) and GluR-interacting protein genes within isolated dentate gyrus granule cells following axotomy of the principal input, the perforant path (PP). Dentate gyrus granule cells were evaluated by microdissection via laser capture microdissection, terminal continuation RNA amplification, and microarray analysis following unilateral PP transections at seven time points. Expression profiles garnered from granule cells on the side ipsilateral to PP transections were compared and contrasted with naive subjects and mice subjected to unilateral occipital cortex lesions. Selected microarray observations were validated by real-time quantitative PCR analysis. Postlesion time-dependent alterations in specific alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors, kainate receptors, N-methyl-D-aspartate (NMDA) receptors, and GluR-interacting protein genes were found across the time course of the study, suggesting a neuroplasticity response associated with the transsynaptic granule cell alterations following axotomy of incoming PP terminals.
Collapse
Affiliation(s)
- Stephen D Ginsberg
- Department of Psychiatry, Center for Dementia Research, Nathan Kline Institute, New York University Langone Medical Center, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA.
| |
Collapse
|
9
|
Khorooshi R, Owens T. Injury-induced type I IFN signaling regulates inflammatory responses in the central nervous system. THE JOURNAL OF IMMUNOLOGY 2010; 185:1258-64. [PMID: 20562259 DOI: 10.4049/jimmunol.0901753] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Innate glial response is critical for the induction of inflammatory mediators and recruitment of leukocytes to sites of the injury in the CNS. We have examined the involvement of type I IFN signaling in the mouse hippocampus following sterile injury (transection of entorhinal afferents). Type I IFNs signal through a receptor (IFNAR), which involves activation of IFN regulatory factor (IRF)9, leading to the induction of IFN-stimulated genes including IRF7, that in turn enhances the induction of type I IFN. Axonal transection induced upregulation of IRF7 and IRF9 in hippocampus. Induction of IRF7 and IRF9 mRNAs was IFNAR dependent. Double-labeling immunofluorescence showed that IRF7 selectively was induced in Mac-1/CD11b(+) macrophages/microglia in hippocampus after axonal transection. IRF7 mRNA was also detected in microglia sorted by flow cytometry. Lack of type I IFN signaling resulted in increased leukocyte infiltration into the lesion-reactive hippocampus. Axonal lesion-induced CXCL10 gene expression was abrogated, whereas matrix metalloproteinase 9 mRNA was elevated in IFNAR-deficient mice. Our findings point to a role for type I IFN signaling in regulation of CNS response to sterile injury.
Collapse
Affiliation(s)
- Reza Khorooshi
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | |
Collapse
|
10
|
Broggini T, Nitsch R, Savaskan NE. Plasticity-related gene 5 (PRG5) induces filopodia and neurite growth and impedes lysophosphatidic acid- and nogo-A-mediated axonal retraction. Mol Biol Cell 2009; 21:521-37. [PMID: 20032306 PMCID: PMC2820418 DOI: 10.1091/mbc.e09-06-0506] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The authors have cloned a novel member of the PRG family that induces filopodia growth in a Cdc42-independent manner. Hence, studies in primary neurons revealed that PRG5 impedes RhoA-mediated axon collapse induced by LPA and Nogo-A. These data reveal a new function of PRG5 with impact on neurite growth in an axonal growth inhibitory environment. Members of the plasticity-related gene (PRG1-4) family are brain-specific integral membrane proteins and implicated in neuronal plasticity, such as filopodia formation and axon growth after brain lesion. Here we report on the cloning of a novel member of the PRG family, PRG5, with high homologies to PRG3. PRG5 is regulated during brain and spinal cord development and is exclusively allocated within the nervous system. When introduced in neurons, PRG5 is distributed in the plasma membrane and induces filopodia as well as axon elongation and growth. Conversely, siRNA mediated knockdown of PRG5 impedes axon growth and disturbs filopodia formation. Here we show that PRG5 induces filopodia growth independently of Cdc42. Moreover, axon collapse and RhoA activation induced by LPA and myelin-associated neurite inhibitor Nogo-A is attenuated in the presence of PRG5, although direct activation of the RhoA-Rho-PIP5K kinase pathway abolishes PRG5 -formed neurites. Thus, we describe here the identification of a novel member of the PRG family that induces filopodia and axon elongation in a Cdc42-independent manner. In addition, PRG5 impedes brain injury-associated growth inhibitory signals upstream of the RhoA-Rho kinase pathway.
Collapse
Affiliation(s)
- Thomas Broggini
- Institute of Cell Biology and Neurobiology, Center of Anatomy, Charité-Universitätsmedizin Berlin, D-10117 Berlin, Germany
| | | | | |
Collapse
|
11
|
Mufson EJ, Counts SE, Perez SE, Ginsberg SD. Cholinergic system during the progression of Alzheimer's disease: therapeutic implications. Expert Rev Neurother 2008; 8:1703-18. [PMID: 18986241 PMCID: PMC2631573 DOI: 10.1586/14737175.8.11.1703] [Citation(s) in RCA: 445] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Alzheimer's disease (AD) is characterized by a progressive phenotypic downregulation of markers within cholinergic basal forebrain (CBF) neurons, frank CBF cell loss and reduced cortical choline acetyltransferase activity associated with cognitive decline. Delaying CBF neurodegeneration or minimizing its consequences is the mechanism of action for most currently available drug treatments for cognitive dysfunction in AD. Growing evidence suggests that imbalances in the expression of NGF, its precursor proNGF and the high (TrkA) and low (p75(NTR)) affinity NGF receptors are crucial factors underlying CBF dysfunction in AD. Drugs that maintain a homeostatic balance between TrkA and p75(NTR) may slow the onset of AD. A NGF gene therapy trial reduced cognitive decline and stimulated cholinergic fiber growth in humans with mild AD. Drugs treating the multiple pathologies and clinical symptoms in AD (e.g., M1 cholinoceptor and/or galaninergic drugs) should be considered for a more comprehensive treatment approach for cholinergic dysfunction.
Collapse
Affiliation(s)
- Elliott J Mufson
- Department of Neurological Sciences, Rush University Medical Center, 1735 W. Harrison, Chicago, IL 60612, USA Tel.: +1 312 563 3570 Fax: +1 312 563 3571
| | - Scott E Counts
- Department of Neurological Sciences, Rush University Medical Center, 1735 W. Harrison, Chicago, IL 60612, USA Tel.: +1 312 563 3570 Fax: +1 312 563 3571
| | - Sylvia E Perez
- Department of Neurological Sciences, Rush University Medical Center, 1735 W. Harrison, Chicago, IL 60612, USA Tel.: +1 312 563 3570 Fax: +1 312 563 3571
| | - Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Departments of Psychiatry, Physiology & Neuroscience, New York University School of Medicine, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA Tel.: +1 845 398 2170 Fax: +1 845 398 5422
| |
Collapse
|
12
|
Yamada M, Shida Y, Takahashi K, Tanioka T, Nakano Y, Tobe T, Yamada M. Prg1 is regulated by the basic helix-loop-helix transcription factor Math2. J Neurochem 2008; 106:2375-84. [PMID: 18643870 DOI: 10.1111/j.1471-4159.2008.05579.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: 11/29/2022]
Abstract
Math2 (NEX-1/NeuroD6) is a member of the basic helix-loop-helix transcription factor family and is involved in neuronal differentiation and maturation. In this study, we identified the genes targeted by Math2 using DNA microarrays and cultured rat cortical cells transfected with Math2. Of the genes regulated by Math2, we focused on plasticity-related gene 1 (Prg1). Prg1 expression induced by Math2 was confirmed in cultured rat cortical cells and PC12 cells analyzed by real-time quantitative PCR. In the promoter region of rat Prg1, we identified four E-boxes [designated -E1 to -E4 (CANNTG)] recognized by the basic helix-loop-helix transcription factor. Using chromatin immunoprecipitation assays, we found that Math2 directly bound to at least one of these E-boxes. The Prg1 reporter assay showed that -E1 was critical for the regulation of Math2-mediated Prg1 expression. Investigation of the functional roles of Math2 and Prg1 in PC12 cells revealed that 72 h after transfection with either Math2 or Prg1, neurite length and number were significantly induced. Co-transfection with Prg1-siRNA completely inhibited Math2-mediated morphological changes. Our results suggest that Math2 directly regulates Prg1 expression and that the Math2-Prg1 cascade plays an important role in neurite outgrowth in PC12 cells.
Collapse
Affiliation(s)
- Misa Yamada
- Department of Psychogeriatrics, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
13
|
Schäfer R, Dehn D, Burbach GJ, Deller T. Differential regulation of chondroitin sulfate proteoglycan mRNAs in the denervated rat fascia dentata after unilateral entorhinal cortex lesion. Neurosci Lett 2008; 439:61-5. [PMID: 18511192 DOI: 10.1016/j.neulet.2008.04.098] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2008] [Revised: 04/29/2008] [Accepted: 04/30/2008] [Indexed: 11/25/2022]
Abstract
Following brain trauma, chondroitin sulphate proteoglycans (CSPGs) are enriched at injury sites and in denervated areas. At injury sites, CSPGs are regarded as inhibitors of axonal regeneration because of their growth inhibitory properties. In areas of denervation their role is less clear, since they are enriched in zones of sprouting, i.e. zones of axonal growth. To identify CSPGs expressed in a denervated brain area and to quantify changes in their mRNA expression, neurocan, brevican, NG2, phosphacan and aggrecan mRNA were analyzed in the rat fascia dentata following entorhinal denervation. Laser microdissection was combined with quantitative RT-PCR to measure mRNA changes specifically within the denervated portion of the molecular layer (1h, 6h, 10h, 12h, 1d, 2d, 3d, 4d, 7d and 14d post-lesion). Changes in glial fibrillary protein mRNA were measured at the same time points and used as lesion control. This approach revealed a differential regulation of CSPG mRNAs in the denervated zone: neurocan, brevican and NG2 mRNA were upregulated with a maximum around 2 days post-lesion. In contrast, aggrecan mRNA levels reached a maximum 7 days post-lesion and phosphacan mRNA levels were not significantly altered. Taken together, our data reveal a temporal pattern in CSPG mRNA expression in the denervated fascia dentata. This suggests specific biological functions for CSPGs during the denervation-induced reorganization process: whereas the early increase in CSPGs in the denervated zone could influence the pattern of sprouting, the late increase of aggrecan mRNA suggests a different role during the late phase of reorganization.
Collapse
Affiliation(s)
- Ruth Schäfer
- Institute of Clinical Neuroanatomy, Johann Wolfgang Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | | | | | | |
Collapse
|
14
|
Deller T, Del Turco D, Rappert A, Bechmann I. Structural reorganization of the dentate gyrus following entorhinal denervation: species differences between rat and mouse. PROGRESS IN BRAIN RESEARCH 2008; 163:501-28. [PMID: 17765735 DOI: 10.1016/s0079-6123(07)63027-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Deafferentation of the dentate gyrus by unilateral entorhinal cortex lesion or unilateral perforant pathway transection is a classical model to study the response of the central nervous system (CNS) to denervation. This model has been extensively characterized in the rat to clarify mechanisms underlying denervation-induced gliosis, transneuronal degeneration of denervated neurons, and collateral sprouting of surviving axons. As a result, candidate molecules have been identified which could regulate these changes, but a causal link between these molecules and the postlesional changes has not yet been demonstrated. To this end, mutant mice are currently studied by many groups. A tacit assumption is that data from the rat can be generalized to the mouse, and fundamental species differences in hippocampal architecture and the fiber systems involved in sprouting are often ignored. In this review, we will (1) provide an overview of some of the basics and technical aspects of the entorhinal denervation model, (2) identify anatomical species differences between rats and mice and will point out their relevance for the axonal reorganization process, (3) describe glial and local inflammatory changes, (4) consider transneuronal changes of denervated dentate neurons and the potential role of reactive glia in this context, and (5) summarize the differences in the reorganization of the dentate gyrus between the two species. Finally, we will discuss the use of the entorhinal denervation model in mutant mice.
Collapse
Affiliation(s)
- Thomas Deller
- Institute of Clinical Neuroanatomy, J.W. Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt/Main, Germany.
| | | | | | | |
Collapse
|
15
|
Savaskan NE, Borchert A, Bräuer AU, Kuhn H. Role for glutathione peroxidase-4 in brain development and neuronal apoptosis: specific induction of enzyme expression in reactive astrocytes following brain injury. Free Radic Biol Med 2007; 43:191-201. [PMID: 17603929 DOI: 10.1016/j.freeradbiomed.2007.03.033] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2007] [Accepted: 03/31/2007] [Indexed: 01/18/2023]
Abstract
Glutathione peroxidase-4 (GPx4) is a multifunctional selenoprotein expressed as mitochondrial, cytosolic, or nuclear isoforms. As a catalytically active enzyme it has been implicated in antioxidative defense, but during sperm development it functions as a structural protein. GPx4 null mice die in utero at midgestation and knockdown of GPx4 during embryogenesis disturbs brain development. To explore the cerebral function of GPx4 we profiled cell-specific enzyme expression at various stages of perinatal brain maturation and investigated its regulation following brain injury by immunohistochemistry, in situ hybridization, and quantitative RT-PCR. Large amounts of GPx4 mRNA were detected in all neuronal layers during perinatal brain development but expression became restricted during postnatal maturation. In adult brain mitochondrial and cytosolic GPx4 isoforms were detected in neurons of cerebral cortex, hippocampus, and cerebellum whereas glial cells were devoid of GPx4. Following selective brain injury expression of the enzyme was upregulated in reactive astrocytes of lesioned areas and deafferented regions but not in neurons. Selective knockdown of GPx4 by small interfering RNA induced depletion of phosphatidylinositol-(4,5)-bisphosphate in the neuronal plasma membrane and subsequently apoptosis as indicated by caspase-3 activation. We hypothesize that astrocytic upregulation of GPx4 in response to injury is part of a protective cascade counteracting further cell damage.
Collapse
Affiliation(s)
- Nicolai E Savaskan
- Division of Cellular Biochemistry, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
16
|
Deller T, Haas CA, Freiman TM, Phinney A, Jucker M, Frotscher M. Lesion-Induced Axonal Sprouting in the Central Nervous System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 557:101-21. [PMID: 16955706 DOI: 10.1007/0-387-30128-3_6] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Injury or neuronal death often come about as a result of brain disorders. Inasmuch as the damaged nerve cells are interconnected via projections to other regions of the brain, such lesions lead to axonal loss in distal target areas. The central nervous system responds to deafferentation by means of plastic remodeling processes, in particular by inducing outgrowth of new axon collaterals from surviving neurons (collateral sprouting). These sprouting processes result in a partial reinnervation, new circuitry, and functional changes within the deafferented brain regions. Lesioning of the entorhinal cortex is an established model system for studying the phenomenon of axonal sprouting. Using this model system, it could be shown that the sprouting process respects the pre-existing lamination pattern of the deafferented fascia dentata, i. e., it is layer-specific. A variety of different molecules are involved in regulating this reorganization process (extracellular matrix molecules, cell adhesion molecules, transcription factors, neurotrophic factors, growth-associated proteins). It is proposed here that molecules of the extracellular matrix define the boundaries of the laminae following entorhinal lesioning and in so doing limit the sprouting process to the deafferented zone. To illustrate the role of axonal sprouting in disease processes, special attention is given to its significance for neurodegenerative disorders, particularly Alzheimer's disease (AD), and temporal lobe epilepsy. Finally, we discuss both the beneficial as well as disadvantageous functional implications of axonal sprouting for the injured organism in question.
Collapse
Affiliation(s)
- Thomas Deller
- Institute of Clinical Neuroanatomy, Johann Wolfgang Goether-University, Frankfurt am Main, Germany
| | | | | | | | | | | |
Collapse
|
17
|
Deller T, Bas Orth C, Vlachos A, Merten T, Del Turco D, Dehn D, Mundel P, Frotscher M. Plasticity of synaptopodin and the spine apparatus organelle in the rat fascia dentata following entorhinal cortex lesion. J Comp Neurol 2006; 499:471-84. [PMID: 16998909 DOI: 10.1002/cne.21103] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Synaptopodin is an actin-associated molecule essential for the formation of a spine apparatus in telencephalic spines. To study whether synaptopodin and the spine apparatus organelle are regulated under conditions of lesion-induced plasticity, synaptopodin and the spine apparatus were analyzed in granule cells of the rat fascia dentata following entorhinal denervation. Confocal microscopy was employed to quantify layer-specific changes in synaptopodin-immunoreactive puncta densities. Electron microscopy was used to quantify layer-specific changes in spine apparatus organelles. Within the denervated middle and outer molecular layers, the layers of deafferentation-induced spine loss, synaptogenesis, and spinogenesis, the density of synaptopodin puncta and the number of spine apparatuses decreased by 4 days postlesion and slowly recovered in parallel with spinogenesis by 180 days postlesion. Within the nondenervated inner molecular layer, the zone without deafferentation-induced spine loss, a rapid loss of synaptopodin puncta and spine apparatuses was also observed. In this layer, spine apparatus densities recovered by 14 days postlesion, in parallel with plastic remodeling at the synaptic level and the postlesional recovery of granule cell activity. These data demonstrate layer-specific changes in the distribution of synaptopodin and the spine apparatus organelle following partial denervation of granule cells: in the layer of spine loss, spine apparatus densities follow spine densities; in the layer of spine maintenance, however, spine apparatus densities appear to be regulated by other signals.
Collapse
Affiliation(s)
- Thomas Deller
- Institute of Clinical Neuroanatomy, J.W. Goethe-University, D-60590 Frankfurt/Main, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Utton MA, Noble WJ, Hill JE, Anderton BH, Hanger DP. Molecular motors implicated in the axonal transport of tau and alpha-synuclein. J Cell Sci 2005; 118:4645-54. [PMID: 16176937 DOI: 10.1242/jcs.02558] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Tau and alpha-synuclein are both proteins implicated in the pathology of neurodegenerative disease. Here we have investigated the mechanisms of axonal transport of tau and alpha-synuclein, because failure of axonal transport has been implicated in the development of several neurodegenerative disorders. We found that the transport of both of these proteins depend on an intact microtubule- but not actin-cytoskeleton, and that tau and alpha-synuclein both move at overall slow rates of transport. We used time-lapse video microscopy to obtain images of live neurons that had been transfected with plasmids expressing proteins tagged with enhanced green fluorescent protein. We found that particulate structures containing tau or alpha-synuclein travel rapidly when moving along axons but spend the majority of the time paused, and these structures have similar characteristics to those previously observed for neurofilaments. The motile particles containing tau or alpha-synuclein colocalise with the fast-transporting molecular motor kinesin-1 in neurons. Co-immunoprecipitation experiments demonstrate that tau and alpha-synuclein are each associated with complexes containing kinesin-1, whereas only alpha-synuclein appears to interact with dynein-containing complexes. In vitro glutathione S-transferase-binding assays using rat brain homogenate or recombinant protein as bait reveals a direct interaction of kinesin-1 light chains 1 and 2 with tau, but not with alpha-synuclein. Our findings suggest that the axonal transport of tau occurs via a mechanism utilising fast transport motors, including the kinesin family of proteins, and that alpha-synuclein transport in neurons may involve both kinesin and dynein motor proteins.
Collapse
Affiliation(s)
- Michelle A Utton
- Department of Neuroscience, King's College London, Institute of Psychiatry, De Crespigny Park, London, SE5 8AF, UK
| | | | | | | | | |
Collapse
|
19
|
Wang Y, Ying GX, Liu X, Wang WY, Dong JH, Ni ZM, Zhou CF. Induction of ephrin-B1 and EphB receptors during denervation-induced plasticity in the adult mouse hippocampus. Eur J Neurosci 2005; 21:2336-46. [PMID: 15932593 DOI: 10.1111/j.1460-9568.2005.04093.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Abstract It has been widely demonstrated that Eph receptors and their ephrin ligands play multiple pivotal roles in the development of the nervous system. However, less is known about their roles in the adult brain. Here we reported the expression of ephrin-B1 and its cognate EphB receptors in the adult mouse hippocampus at 3, 7, 15, 30 and 60 days after transections of the entorhinal afferents. In situ hybridization and immunohistochemistry showed the time-dependent up-regulation of ephrin-B1 in the denervated areas of the hippocampus, which initiated at 3 days postlesion (dpl), reached maximal levels at 7-15 dpl, remained slightly elevated at 30 dpl and recovered to normal levels by 60 dpl. Double labeling of ephrin-B1 and glial fibrillary acidic protein revealed that ephrin-B1-expressing cells in the denervated areas were reactive astrocytes. Furthermore, a ligand-binding assay using ephrin-B1/Fc chimera protein also displayed the up-regulation of EphB receptors in the denervated areas of the hippocampus in a similar manner to that of ephrin-B1. Within the first week postlesion, the EphB receptors were expressed by reactive astrocytes. After 7 dpl, however, EphB receptors were expressed not only by reactive astrocytes but also first by sprouting axons and later by regrowing dendrites. These results suggest that the ephrin-B1/EphB system may participate in the lesion-induced plasticity processes in the adult mouse hippocampus.
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of Neurobiology, Shanghai Institute of Physiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, PR China
| | | | | | | | | | | | | |
Collapse
|
20
|
Wang Y, Ni ZM, Zhou CF. Denervation-induced spatiotemporal upregulation of ephrin-A2 in the mouse hippocampus after transections of the perforant path. FEBS Lett 2005; 579:1055-60. [PMID: 15710390 DOI: 10.1016/j.febslet.2005.01.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2004] [Revised: 12/23/2004] [Accepted: 01/03/2005] [Indexed: 10/25/2022]
Abstract
Transections of the entorhinal afferent fibers to hippocampus, perforant path (PP), result in the denervation in specific hippocampal subregions, which is followed by a series of plastic events including axon sprouting and reactive synaptogenesis. Many growth-associated molecules are thought to participate in these events. In the present study, we proved the upregulation of ephrin-A2 in the denervated areas of the ipsilateral hippocampus following PP transections. Interestingly, when the elevation of ephrin-A2 reached the maximum axon sprouting in the denervated areas almost finished, implying the possible inhibitory effect of ephrin-A2 on sprouting. In addition, ephrin-A2 expression was observed in synapses during reactive synaptogenesis, suggesting that this molecule might also be implicated in the formation and maturation of synapses in the denervated areas.
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of Neurobiology, Shanghai Institute of Physiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, PR China
| | | | | |
Collapse
|
21
|
Hofmann F, Guenther E, Hämmerle H, Leibrock C, Berezin V, Bock E, Volkmer H. Functional re-establishment of the perforant pathway in organotypic co-cultures on microelectrode arrays. Brain Res 2004; 1017:184-96. [PMID: 15261114 DOI: 10.1016/j.brainres.2004.05.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2004] [Indexed: 11/24/2022]
Abstract
Co-cultures of entorhinal cortex (EC) and dentate gyrus (DG) explants are a useful model system to study the formation and stabilization of axonal projections. We adapted this model system to EC-DG co-cultures on microelectrode arrays (MEA) for the characterization of axonal projections on a functional level for days and weeks. EC and DG explants were placed on MEA to allow for the reconstitution of perforant pathway projections. Connections formed were characterized by morphological and electrophysiological analyses to verify characteristic features of perforant pathway signal transmission. Morphological analysis reveals proper projection of EC neurons into the molecular layer of the DG. Examination of synaptic transmission after high frequency stimulation imply unidirectional connections that used glutamate receptors of the AMPA/kainate type as main mediators of excitatory signal transmission. The system was evaluated by the introduction of the NCAM binding peptide C3d. In accordance with in vivo and in vitro experiments C3d modulated signal transmission by NCAM-related mechanisms resulting in morphological re-arrangements.
Collapse
Affiliation(s)
- Frank Hofmann
- Naturwissenschaftliches und Medizinisches Institut an der Universität Tübingen, Markwiesenstr. 55, D-72770, Reutlingen, Germany
| | | | | | | | | | | | | |
Collapse
|
22
|
Phinney AL, Calhoun ME, Woods AG, Deller T, Jucker M. Stereological analysis of the reorganization of the dentate gyrus following entorhinal cortex lesion in mice. Eur J Neurosci 2004; 19:1731-40. [PMID: 15078547 DOI: 10.1111/j.1460-9568.2004.03280.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Denervation of the dentate gyrus by entorhinal cortex lesion has been widely used to study the reorganization of neuronal circuits following central nervous system lesion. Expansion of the non-denervated inner molecular layer (commissural/associational zone) of the dentate gyrus and increased acetylcholinesterase-positive fibre density in the denervated outer molecular layer have commonly been regarded as markers for sprouting following entorhinal cortex lesion. However, because this lesion extensively denervates the outer molecular layer and causes tissue shrinkage, stereological analysis is required for an accurate evaluation of sprouting. To this end we have performed unilateral entorhinal cortex lesions in adult C57BL/6J mice and have assessed atrophy and sprouting in the dentate gyrus using modern unbiased stereological techniques. Results revealed the expected increases in commissural/associational zone width and density of acetylcholinesterase-positive fibres on single brain sections. Yet, stereological analysis failed to demonstrate concomitant increases in layer volume or total acetylcholinesterase-positive fibre length. Interestingly, calretinin-positive fibres did grow beyond the border of the commissural/associational zone into the denervated layer and were regarded as sprouting axons. Thus, our data suggest that in C57BL/6J mice shrinkage of the hippocampus rather than growth of fibres underlies the two morphological phenomena most often cited as evidence of regenerative sprouting following entorhinal cortex lesion. Moreover, our data suggest that regenerative axonal sprouting in the mouse dentate gyrus following entorhinal cortex lesion may be best assessed at the single-fibre level.
Collapse
Affiliation(s)
- A L Phinney
- Neuropathology Laboratory, Institute of Pathology, University of Basel, Basel, Switzerland
| | | | | | | | | |
Collapse
|
23
|
Bräuer AU, Nitsch R, Savaskan NE. Identification of macrophage/microglia activation factor (MAF) associated with late endosomes/lysosomes in microglial cells. FEBS Lett 2004; 563:41-8. [PMID: 15063720 DOI: 10.1016/s0014-5793(04)00244-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2004] [Revised: 02/20/2004] [Accepted: 02/25/2004] [Indexed: 11/22/2022]
Abstract
Damage to the central nervous system triggers rapid activation and specific migration of glial cells towards the lesion site. There, glial cells contribute heavily to secondary neuronal changes that take place after lesion. In an attempt to identify the molecular cues of glial activation following brain trauma we performed differential display reverse transcription-polymerase chain reaction screenings from lesioned and control hippocampus. Here we report on the identification of the macrophage/microglia activation factor (MAF), a new membrane protein with seven putative transmembrane domains. Expression analysis revealed that MAF is predominantly expressed in microglial cells in the brain, and is upregulated following brain lesion. Overexpression of MAF in non-glial cells shows an intracellular codistribution with the lysosomal marker endosome/lysosome-associated membrane protein-1 (lamp-1). Furthermore, MAF-transfected cells show that MAF is primarily associated with late endosomes/lysosomes, and that this association can be disrupted by activation of protein kinase C-dependent pathways. In conclusion, these results imply that MAF is involved in the dynamics of lysosomal membranes associated with microglial activation following brain lesion.
Collapse
Affiliation(s)
- Anja U Bräuer
- Institute of Cell Biology and Neurobiology, Center for Anatomy, Charité University Medical School Berlin, Philippstr. 12, D-10115 Berlin, Germany
| | | | | |
Collapse
|
24
|
Del Turco D, Woods AG, Gebhardt C, Phinney AL, Jucker M, Frotscher M, Deller T. Comparison of commissural sprouting in the mouse and rat fascia dentata after entorhinal cortex lesion. Hippocampus 2003; 13:685-99. [PMID: 12962314 DOI: 10.1002/hipo.10118] [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: 11/12/2022]
Abstract
Reactive axonal sprouting occurs in the fascia dentata after entorhinal cortex lesion. This sprouting process has been described extensively in the rat, and plasticity-associated molecules have been identified that might be involved in its regulation. To demonstrate causal relationships between these candidate molecules and the axonal reorganization process, it is reasonable to analyze knockout and transgenic animals after entorhinal cortex lesion, and because gene knockouts are primarily generated in mice, it is necessary to characterize the sprouting response after entorhinal cortex lesion in this species. In the present study, Phaseolus vulgaris-leucoagglutinin (PHAL) tracing was used to analyze the commissural projection to the inner molecular layer in mice with longstanding entorhinal lesions. Because the commissural projection to the fascia dentata is neurochemically heterogeneous, PHAL tracing was combined with immunocytochemistry for calretinin, a marker for commissural/associational mossy cell axons. Using both techniques singly as well as in combination (double-immunofluorescence) at the light or electron microscopic level, it could be shown that in response to entorhinal lesion mossy cell axons leave the main commissural fiber plexus, invade the denervated middle molecular layer, and form asymmetric synapses within the denervated zone. Thus, the commissural sprouting response in mice has a considerable translaminar component. This is in contrast to the layer-specific commissural sprouting observed in rats, in which the overwhelming majority of mossy cell axons remain within their home territory. These data demonstrate an important species difference in the commissural/associational sprouting response between rats and mice that needs to be taken into account in future studies.
Collapse
Affiliation(s)
- Domenico Del Turco
- Institute of Clinical Neuroanatomy, J. W. Goethe University, Frankfurt/Main, Germany
| | | | | | | | | | | | | |
Collapse
|
25
|
Bräuer AU, Savaskan NE, Plaschke M, Ninnemann O, Nitsch R. Cholecystokinin expression after hippocampal deafferentiation: molecular evidence revealed by differential display-reverse transcription-polymerase chain reaction. Neuroscience 2003; 121:111-21. [PMID: 12946704 DOI: 10.1016/s0306-4522(03)00336-1] [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: 11/17/2022]
Abstract
The cortical information flow via the perforant path represents a major excitatory projection to the hippocampus. Lesioning this projection leads to massive degeneration and subsequently to reorganization in its termination zones as well as in primary non-affected subfields of the hippocampus. The molecular mechanisms and factors which are involved in the postlesional events are poorly defined. Using a differential display reverse transcription-polymerase chain reaction (DDRT-PCR) strategy, we located one band which occurred only in control hippocampus lanes and almost disappeared in the lanes of lesioned hippocampi. By sequencing, we identified the corresponding gene as cholecystokinin (CCK). Northern blot analysis confirmed a decreased transcription of CCK after lesion. In situ hybridization analysis was performed for localization and quantification of altered CCK transcription. We noted a significant downregulation of CCK transcription in the hippocampus (20%) and in the contralateral cortex (12%) 1-day after lesion (dal) and an increased signal in the ipsilateral cortex (10.5%). This pattern was altered, showing upregulation of CCK mRNA expression, reaching its highest level of 70% above control levels at 5 dal. In the hippocampus, the control level was reached again at 21 dal, whereas the cortex reached the control level at 10 dal. In comparison, the mRNA transcripts of the receptors CCK(A) and CCK(B) remained unchanged. Since CCK-containing neurons are involved in the modulation of pyramidal and granule cell excitability, our data indicate a time course correlation between CCK mRNA expression and postlesional axonal sprouting response in the hippocampus.
Collapse
Affiliation(s)
- A U Bräuer
- Department of Cell and Neurobiology, Oskar-Hertwig House, Humboldt University Medical School Charité, Philippstrasse 12, Philippstrasse 12, D-10115 Berlin, Germany.
| | | | | | | | | |
Collapse
|
26
|
Roysommuti S, Carroll SL, Wyss JM. Neuregulin-1β modulates in vivo entorhinal–hippocampal synaptic transmission in adult rats. Neuroscience 2003; 121:779-85. [PMID: 14568036 DOI: 10.1016/s0306-4522(03)00503-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Neuregulin-1 (NRG-1) proteins and their erbB receptors are essential for neuronal development during embryogenesis and may contribute importantly to neuronal function in the adult brain. This study tests the hypothesis that NRG-1beta acts as a modulator of synaptic activity in the adult brain, specifically at hippocampal formation synapses. Adult, male Sprague-Dawley rats were anesthetized and a recording electrode with an attached stainless steel microinjector was stereotaxically positioned to record field potentials (fEPSP) in either the dentate gyrus or the cornu ammonis (CA) 1 field of the hippocampus. The entorhinal cortex was continuously stimulated via a paired stainless steel electrode. Microinjection of NRG-1beta significantly increased the slope of the fEPSP in the dentate gyrus in a dose-dependent manner. Compared with a low dose (20 nM), a high dose (100 nM) of NRG-1beta induced a shorter latency response that was of greater magnitude. Responses to NRG-1beta were abolished by pretreatment with a selective, reversible erbB tyrosine kinase inhibitor, PD158780 (100 microM). Further, PD158780 (100 microM) itself significantly decreased the entorhinal-dentate fESPS slope by about 15%. Neither equimolar (100 nM) nor hypermolar (100 microM) sucrose or heat-inactivated NRG-1beta (100 nM) significantly altered the entorhinal-dentate fEPSP slope. In contrast to its effect at the entorhinal-dentate synapse, NRG-1beta (100 nM) depressed, and PD158780 potentiated entorhinal-CA1 synaptic transmission. Thus, in adult rats NRG-1beta potentiates transmission at the entorhinal-dentate synapse but suppresses transmission at the entorhinal-CA1 synapse. These observations indicate that NRG-1 is not only a developmental growth factor, but also modifies synaptic transmission in adult rat brain.
Collapse
Affiliation(s)
- S Roysommuti
- Department of Cell Biology, 1900 University Boulevard, THT 950, University of Alabama at Birmingham, 35294-0006, USA
| | | | | |
Collapse
|
27
|
Bushong EA, Martone ME, Ellisman MH. Examination of the relationship between astrocyte morphology and laminar boundaries in the molecular layer of adult dentate gyrus. J Comp Neurol 2003; 462:241-51. [PMID: 12794746 DOI: 10.1002/cne.10728] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Astrocytes are known to play an integral role in the development of compartmental boundaries in the brain and in the creation of trauma-induced boundaries. However, the physical relationship between astrocytes and such boundaries in the adult brain is less clear. If astrocytes do respect or play an ongoing role in maintaining such boundaries, a correlation between the position of such a boundary and the morphology of neighboring astrocytes might be observable. In this study, we examined the distribution of astrocytes with respect to the laminar boundaries compartmentalizing afferents to the dentate gyrus molecular layer. In addition, we attempted to determine whether astrocyte morphology is influenced by these laminar boundaries. To this end, protoplasmic astrocytes in the adult rat dentate gyrus were revealed with fluorescent tracer dyes and subsequently analyzed with respect to laminar boundaries demarcated by means of immunolabeling for the lamina-specific molecules EphA4 and neural cell adhesion molecule (N-CAM). We find that astrocyte distribution is influenced by the boundary separating the associational/commissural and perforant path afferents. In addition, we show that astrocytes in this region are polarized in their morphology, unlike typically stellate astrocytes, but that the laminar boundaries themselves do not appear to confer this morphology. This polarized morphology, however acquired, may have import for the functioning of astrocytes within the highly organized composition of the dentate gyrus molecular layer and for the overall microphysiology of this and other brain regions.
Collapse
Affiliation(s)
- Eric A Bushong
- National Center for Microscopy and Imaging Research, University of California- San Diego, La Jolla, California 92093, USA
| | | | | |
Collapse
|
28
|
Prang P, Del Turco D, Deller T. Associational sprouting in the mouse fascia dentata after entorhinal lesion in vitro. Brain Res 2003; 978:205-12. [PMID: 12834915 DOI: 10.1016/s0006-8993(03)02836-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Collateral sprouting is a form of neuronal plasticity observed in brain following injury. In order to establish an in vitro model of collateral sprouting, entorhino-hippocampal slice cultures were prepared from brain of C57BL/6 mouse pups (P1-4) and incubated for 14-16 days in vitro. Thereafter, entorhino-hippocampal fibers were cut and the outer molecular layer of the fascia dentata was denervated. At this age, entorhino-hippocampal fibers do not regenerate, as could be shown using anterograde tracing with Miniruby. Sprouting of associational mossy cell axons was monitored using calretinin-immunocytochemistry. Control and lesioned entorhino-hippocampal slices were studied at 1, 5, and 10 days postlesion. Whereas only the inner portion of the molecular layer was occupied by calretinin-positive mossy cell axons in controls and after 1 and 5 days postlesion, the entire width of the molecular layer was occupied by associational fibers by 10 days postlesion. Thus, robust sprouting of associational mossy cell axons occurs in response to entorhinal denervation in vitro. Using organotypic entorhino-hippocampal slices of genetically engineered mice, this sprouting model can be used to identify molecules involved in the regulation of sprouting following brain injury.
Collapse
Affiliation(s)
- Peter Prang
- Institute of Clinical Neuroanatomy, JW Goethe University, Theodor-Stern-Kai 7, D-60590, Frankfurt/Main, Germany
| | | | | |
Collapse
|
29
|
Bräuer AU, Savaskan NE, Kühn H, Prehn S, Ninnemann O, Nitsch R. A new phospholipid phosphatase, PRG-1, is involved in axon growth and regenerative sprouting. Nat Neurosci 2003; 6:572-8. [PMID: 12730698 DOI: 10.1038/nn1052] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2003] [Accepted: 02/28/2003] [Indexed: 11/09/2022]
Abstract
Outgrowth of axons in the central nervous system is governed by specific molecular cues. Molecules detected so far act as ligands that bind to specific receptors. Here, we report a new membrane-associated lipid phosphate phosphatase that we have named plasticity-related gene 1 (PRG-1), which facilitates axonal outgrowth during development and regenerative sprouting. PRG-1 is specifically expressed in neurons and is located in the membranes of outgrowing axons. There, it acts as an ecto-enzyme and attenuates phospholipid-induced axon collapse in neurons and facilitates outgrowth in the hippocampus. Thus, we propose a novel mechanism by which axons are able to control phospholipid-mediated signaling and overcome the growth-inhibiting, phospholipid-rich environment of the extracellular space.
Collapse
Affiliation(s)
- Anja U Bräuer
- Institute of Anatomy, Department of Cell Biology and Neurobiology, Philippstr. 12, Humboldt University Medical School Charité, D-10115 Berlin, Germany
| | | | | | | | | | | |
Collapse
|
30
|
Stamer K, Vogel R, Thies E, Mandelkow E, Mandelkow EM. Tau blocks traffic of organelles, neurofilaments, and APP vesicles in neurons and enhances oxidative stress. J Cell Biol 2002; 156:1051-63. [PMID: 11901170 PMCID: PMC2173473 DOI: 10.1083/jcb.200108057] [Citation(s) in RCA: 678] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We studied the effect of microtubule-associated tau protein on trafficking of vesicles and organelles in primary cortical neurons, retinal ganglion cells, and neuroblastoma cells. Tau inhibits kinesin-dependent transport of peroxisomes, neurofilaments, and Golgi-derived vesicles into neurites. Loss of peroxisomes makes cells vulnerable to oxidative stress and leads to degeneration. In particular, tau inhibits transport of amyloid precursor protein (APP) into axons and dendrites, causing its accumulation in the cell body. APP tagged with yellow fluorescent protein and transfected by adenovirus associates with vesicles moving rapidly forward in the axon (approximately 80%) and slowly back (approximately 20%). Both movements are strongly inhibited by cotransfection with fluorescently tagged tau (cyan fluorescent protein-tau) as seen by two-color confocal microscopy. The data suggests a linkage between tau and APP trafficking, which may be significant in Alzheimer's disease.
Collapse
Affiliation(s)
- K Stamer
- Max-Planck-Unit for Structural Molecular Biology, 22607 Hamburg, Germany
| | | | | | | | | |
Collapse
|