1
|
Smith RL, Sawiak SJ, Dorfschmidt L, Dutcher EG, Jones JA, Hahn JD, Sporns O, Swanson LW, Taylor PA, Glen DR, Dalley JW, McMahon FJ, Raznahan A, Vértes PE, Bullmore ET. Development and early life stress sensitivity of the rat cortical microstructural similarity network. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.20.629759. [PMID: 39803427 PMCID: PMC11722359 DOI: 10.1101/2024.12.20.629759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
The rat offers a uniquely valuable animal model in neuroscience, but we currently lack an individual-level understanding of the in vivo rat brain network. Here, leveraging longitudinal measures of cortical magnetization transfer ratio (MTR) from in vivo neuroimaging between postnatal days 20 (weanling) and 290 (mid-adulthood), we design and implement a computational pipeline that captures the network of structural similarity (MIND, morphometric inverse divergence) between each of 53 distinct cortical areas. We first characterized the normative development of the network in a cohort of rats undergoing typical development (N=47), and then contrasted these findings with a cohort exposed to early life stress (ELS, N=40). MIND as a metric of cortical similarity and connectivity was validated by cortical cytoarchitectonics and axonal tract-tracing data. The normative rat MIND network had high between-study reliability and complex topological properties including a rich club. Similarity changed during post-natal and adolescent development, including a phase of fronto-hippocampal convergence, or increasing inter-areal similarity. An inverse process of increasing fronto-hippocampal dissimilarity was seen with post-adult aging. Exposure to ELS in the form of maternal separation appeared to accelerate the normative trajectory of brain development - highlighting embedding of stress in the dynamic rat brain network. Our work provides novel tools for systems-level study of the rat brain that can now be used to understand network-based underpinnings of complex lifespan behaviors and experimental manipulations that this model organism allows.
Collapse
Affiliation(s)
- Rachel L. Smith
- Department of Psychiatry, University of Cambridge, Cambridge, CB2 0SZ, UK
- Human Genetics Branch, National Institute of Mental Health, Bethesda, MD, USA 20892
| | - Stephen J. Sawiak
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Site, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EL, UK
| | - Lena Dorfschmidt
- Department of Psychiatry, University of Cambridge, Cambridge, CB2 0SZ, UK
| | - Ethan G. Dutcher
- Department of Psychology, University of Cambridge, Cambridge, CB2 3EB, UK
| | - Jolyon A. Jones
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Site, Cambridge, UK
- Department of Psychology, University of Cambridge, Cambridge, CB2 3EB, UK
| | - Joel D. Hahn
- Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA 90089
| | - Olaf Sporns
- Indiana University Network Science Institute, Indiana University, Bloomington, IN, USA 47405
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA 47405
| | - Larry W. Swanson
- Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA 90089
| | - Paul A. Taylor
- Scientific and Statistical Computing Core, National Institute of Mental Health, NIH, Bethesda, MD, USA 20892
| | - Daniel R. Glen
- Scientific and Statistical Computing Core, National Institute of Mental Health, NIH, Bethesda, MD, USA 20892
| | - Jeffrey W. Dalley
- Department of Psychiatry, University of Cambridge, Cambridge, CB2 0SZ, UK
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Site, Cambridge, UK
- Department of Psychology, University of Cambridge, Cambridge, CB2 3EB, UK
| | - Francis J. McMahon
- Human Genetics Branch, National Institute of Mental Health, Bethesda, MD, USA 20892
| | - Armin Raznahan
- Human Genetics Branch, National Institute of Mental Health, Bethesda, MD, USA 20892
| | - Petra E. Vértes
- Department of Psychiatry, University of Cambridge, Cambridge, CB2 0SZ, UK
| | - Edward T. Bullmore
- Department of Psychiatry, University of Cambridge, Cambridge, CB2 0SZ, UK
| |
Collapse
|
2
|
Kamen Y, Chapman TW, Piedra ET, Ciolkowski ME, Hill RA. Transient upregulation of procaspase-3 during oligodendrocyte fate decisions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.623446. [PMID: 39605489 PMCID: PMC11601457 DOI: 10.1101/2024.11.13.623446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Oligodendrocytes are generated throughout life and in neurodegenerative conditions from brain resident oligodendrocyte precursor cells (OPCs). The transition from OPC to oligodendrocyte involves a complex cascade of molecular and morphological states that position the cell to make a fate decision to integrate as a myelinating oligodendrocyte or die through apoptosis. Oligodendrocyte maturation impacts the cell death mechanisms that occur in degenerative conditions, but it is unclear if and how the cell death machinery changes as OPCs transition into oligodendrocytes. Here, we discovered that differentiating oligodendrocytes transiently upregulate the zymogen procaspase-3, equipping these cells to make a survival decision during differentiation. Pharmacological inhibition of caspase-3 decreases oligodendrocyte density, indicating that procaspase-3 upregulation promotes differentiation. Moreover, using procaspase-3 as a marker, we show that oligodendrocyte differentiation continues in the aging cortex and white matter. Taken together, our data establish procaspase-3 as a differentiating oligodendrocyte marker and provide insight into the underlying mechanisms occurring during the decision to integrate or die.
Collapse
Affiliation(s)
- Yasmine Kamen
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Timothy W. Chapman
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Enrique T. Piedra
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | | | - Robert A. Hill
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|
3
|
Janowska J, Gargas J, Zajdel K, Wieteska M, Lipinski K, Ziemka‐Nalecz M, Frontczak‐Baniewicz M, Sypecka J. Oligodendrocyte progenitor cells' fate after neonatal asphyxia-Puzzling implications for the development of hypoxic-ischemic encephalopathy. Brain Pathol 2024; 34:e13255. [PMID: 38504469 PMCID: PMC11483519 DOI: 10.1111/bpa.13255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 03/01/2024] [Indexed: 03/21/2024] Open
Abstract
Premature birth or complications during labor can cause temporary disruption of cerebral blood flow, often followed by long-term disturbances in brain development called hypoxic-ischemic (HI) encephalopathy. Diffuse damage to the white matter is the most frequently detected pathology in this condition. We hypothesized that oligodendrocyte progenitor cell (OPC) differentiation disturbed by mild neonatal asphyxia may affect the viability, maturation, and physiological functioning of oligodendrocytes. To address this issue, we studied the effect of temporal HI in the in vivo model in P7 rats with magnetic resonance imaging (MRI), microscopy techniques and biochemical analyses. Moreover, we recreated the injury in vitro performing the procedure of oxygen-glucose deprivation on rat neonatal OPCs to determine its effect on cell viability, proliferation, and differentiation. In the in vivo model, MRI evaluation revealed changes in the volume of different brain regions, as well as changes in the directional diffusivity of water in brain tissue that may suggest pathological changes to myelinated neuronal fibers. Hypomyelination was observed in the cortex, striatum, and CA3 region of the hippocampus. Severe changes to myelin ultrastructure were observed, including delamination of myelin sheets. Interestingly, shortly after the injury, an increase in oligodendrocyte proliferation was observed, followed by an overproduction of myelin proteins 4 weeks after HI. Results verified with the in vitro model indicate, that in the first days after damage, OPCs do not show reduced viability, intensively proliferate, and overexpress myelin proteins and oligodendrocyte-specific transcription factors. In conclusion, despite the increase in oligodendrocyte proliferation and myelin protein expression after HI, the production of functional myelin sheaths in brain tissue is impaired. Presented study provides a detailed description of oligodendrocyte pathophysiology developed in an effect of HI injury, resulting in an altered CNS myelination. The described models may serve as useful tools for searching and testing effective of effective myelination-supporting therapies for HI injuries.
Collapse
Affiliation(s)
- Justyna Janowska
- Department of NeuroRepairMossakowski Medical Research Institute PASWarsawPoland
| | - Justyna Gargas
- Department of NeuroRepairMossakowski Medical Research Institute PASWarsawPoland
| | - Karolina Zajdel
- NOMATEN Center of Excellence, National Center for Nuclear ResearchOtwockPoland
- Electron Microscopy Research UnitMossakowski Medical Research Institute PASWarsawPoland
| | - Michal Wieteska
- Small Animal Magnetic Resonance Imaging LaboratoryMossakowski Medical Research Institute PASWarsawPoland
| | - Kamil Lipinski
- Division of Nuclear and Medical ElectronicsWarsaw University of TechnologyWarsawPoland
| | | | | | - Joanna Sypecka
- Department of NeuroRepairMossakowski Medical Research Institute PASWarsawPoland
| |
Collapse
|
4
|
Rajani RM, Ellingford R, Hellmuth M, Harris SS, Taso OS, Graykowski D, Lam FKW, Arber C, Fertan E, Danial JSH, Swire M, Lloyd M, Giovannucci TA, Bourdenx M, Klenerman D, Vassar R, Wray S, Sala Frigerio C, Busche MA. Selective suppression of oligodendrocyte-derived amyloid beta rescues neuronal dysfunction in Alzheimer's disease. PLoS Biol 2024; 22:e3002727. [PMID: 39042667 PMCID: PMC11265669 DOI: 10.1371/journal.pbio.3002727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/25/2024] Open
Abstract
Reduction of amyloid beta (Aβ) has been shown to be effective in treating Alzheimer's disease (AD), but the underlying assumption that neurons are the main source of pathogenic Aβ is untested. Here, we challenge this prevailing belief by demonstrating that oligodendrocytes are an important source of Aβ in the human brain and play a key role in promoting abnormal neuronal hyperactivity in an AD knock-in mouse model. We show that selectively suppressing oligodendrocyte Aβ production improves AD brain pathology and restores neuronal function in the mouse model in vivo. Our findings suggest that targeting oligodendrocyte Aβ production could be a promising therapeutic strategy for treating AD.
Collapse
Affiliation(s)
- Rikesh M. Rajani
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - Robert Ellingford
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - Mariam Hellmuth
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - Samuel S. Harris
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - Orjona S. Taso
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - David Graykowski
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - Francesca Kar Wey Lam
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - Charles Arber
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London, United Kingdom
| | - Emre Fertan
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
- UK Dementia Research Institute at University of Cambridge, Cambridge, United Kingdom
| | - John S. H. Danial
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
- UK Dementia Research Institute at University of Cambridge, Cambridge, United Kingdom
- School of Physics and Astronomy, University of St Andrews, St. Andrews, United Kingdom
| | - Matthew Swire
- Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Marcus Lloyd
- Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Tatiana A. Giovannucci
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London, United Kingdom
| | - Mathieu Bourdenx
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
- UK Dementia Research Institute at University of Cambridge, Cambridge, United Kingdom
| | - Robert Vassar
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Selina Wray
- Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London, United Kingdom
| | - Carlo Sala Frigerio
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - Marc Aurel Busche
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| |
Collapse
|
5
|
Arreguin AJ, Shao Z, Colognato H. Dmd mdx mice have defective oligodendrogenesis, delayed myelin compaction and persistent hypomyelination. Dis Model Mech 2024; 17:dmm050115. [PMID: 38721692 PMCID: PMC11095635 DOI: 10.1242/dmm.050115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 03/28/2024] [Indexed: 05/18/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by mutations in the DMD gene, resulting in the loss of dystrophin, a large cytosolic protein that links the cytoskeleton to extracellular matrix receptors in skeletal muscle. Aside from progressive muscle damage, many patients with DMD also have neurological deficits of unknown etiology. To investigate potential mechanisms for DMD neurological deficits, we assessed postnatal oligodendrogenesis and myelination in the Dmdmdx mouse model. In the ventricular-subventricular zone (V-SVZ) stem cell niche, we found that oligodendrocyte progenitor cell (OPC) production was deficient, with reduced OPC densities and proliferation, despite a normal stem cell niche organization. In the Dmdmdx corpus callosum, a large white matter tract adjacent to the V-SVZ, we also observed reduced OPC proliferation and fewer oligodendrocytes. Transmission electron microscopy further revealed significantly thinner myelin, an increased number of abnormal myelin structures and delayed myelin compaction, with hypomyelination persisting into adulthood. Our findings reveal alterations in oligodendrocyte development and myelination that support the hypothesis that changes in diffusion tensor imaging seen in patients with DMD reflect developmental changes in myelin architecture.
Collapse
Affiliation(s)
- Andrea J. Arreguin
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
- Medical Scientist Training Program (MSTP), Stony Brook University, Stony Brook, NY 11794-8651, USA
| | - Zijian Shao
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| | - Holly Colognato
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
6
|
Gakare SG, Bhatt JM, Narasimhan KKS, Dravid SM. Glutamate delta-1 receptor regulates oligodendrocyte progenitor cell differentiation and myelination in normal and demyelinating conditions. PLoS One 2023; 18:e0294583. [PMID: 37983226 PMCID: PMC10659214 DOI: 10.1371/journal.pone.0294583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/05/2023] [Indexed: 11/22/2023] Open
Abstract
In this study, we investigated the role of glutamate delta 1 receptor (GluD1) in oligodendrocyte progenitor cell (OPC)-mediated myelination during basal (development) and pathophysiological (cuprizone-induced demyelination) conditions. Initially, we sought to determine the expression pattern of GluD1 in OPCs and found a significant colocalization of GluD1 puncta with neuron-glial antigen 2 (NG2, OPC marker) in the motor cortex and dorsal striatum. Importantly, we found that the ablation of GluD1 led to an increase in the number of myelin-associated glycoprotein (MAG+) cells in the corpus callosum and motor cortex at P40 without affecting the number of NG2+ OPCs, suggesting that GluD1 loss selectively facilitates OPC differentiation rather than proliferation. Further, deletion of GluD1 enhanced myelination in the corpus callosum and motor cortex, as indicated by increased myelin basic protein (MBP) staining at P40, suggesting that GluD1 may play an essential role in the developmental regulation of myelination during the critical window period. In contrast, in cuprizone-induced demyelination, we observed reduced MBP staining in the corpus callosum of GluD1 KO mice. Furthermore, cuprizone-fed GluD1 KO mice showed more robust motor deficits. Collectively, our results demonstrate that GluD1 plays a critical role in OPC regulation and myelination in normal and demyelinating conditions.
Collapse
Affiliation(s)
- Sukanya G. Gakare
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, United States of America
| | - Jay M. Bhatt
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, United States of America
| | - Kishore Kumar S. Narasimhan
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, United States of America
| | - Shashank M. Dravid
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, United States of America
| |
Collapse
|
7
|
A Disintegrin and Metalloproteinase 10 (ADAM10) Is Essential for Oligodendrocyte Precursor Development and Myelination in the Mouse Brain. Mol Neurobiol 2023; 60:1675-1689. [PMID: 36550333 PMCID: PMC9899191 DOI: 10.1007/s12035-022-03163-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
A disintegrin and metalloproteinase 10 (ADAM10) plays an essential role in the regulation of survival, proliferation, migration, and differentiation of various neural cells. Nevertheless, the role of ADAM10 in oligodendrocyte precursors (OPCs) and myelination in the central nervous system (CNS) of developing and adult mouse brains is still unknown. We generated ADAM10 conditional knockout (ADAM10 cKO) mice lacking the ADAM10 gene primarily in OPCs by crossing NG2-Cre mice with ADAM10 loxp/loxp mice. We found that OPCs expressed ADAM10 in the mouse corpus callosum and the hippocampus. ADAM10 cKO mice showed significant loss of back hair and reduction in weight and length on postnatal (30 ± 2.1) day, died at (65 ± 5) days after birth, and exhibited the "anxiety and depression-like" performances. Conditional knockout of ADAM10 in OPCs resulted in a prominent increase in myelination and a decrease in the number of OPCs in the corpus callosum at P30 owing to premyelination and lack of proliferation of OPCs. Moreover, the number of proliferating OPCs and mature oligodendrocytes (OLs) also decreased with age in the corpus callosum of ADAM10 cKO mice from P30 to P60. Western blot and RT-PCR results showed that the activation of Notch-1 and its four target genes, Hes1, Hes5, Hey1, and Hey2, was inhibited in the corpus callosum tissue of ADAM10 knockout mice. In our study, we provided experimental evidence to demonstrate that ADAM10 is essential for modulating CNS myelination and OPC development by activating Notch-1 signaling in the developing and adult mouse brain.
Collapse
|
8
|
Kurki SN, Uvarov P, Pospelov AS, Trontti K, Hübner AK, Srinivasan R, Watanabe M, Hovatta I, Hübner CA, Kaila K, Virtanen MA. Expression patterns of NKCC1 in neurons and non-neuronal cells during cortico-hippocampal development. Cereb Cortex 2022; 33:5906-5923. [PMID: 36573432 PMCID: PMC10183754 DOI: 10.1093/cercor/bhac470] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/05/2022] [Accepted: 11/06/2022] [Indexed: 12/28/2022] Open
Abstract
Abstract
The Na-K-2Cl cotransporter NKCC1 is widely expressed in cells within and outside the brain. However, our understanding of its roles in brain functions throughout development, as well as in neuropsychiatric and neurological disorders, has been severely hindered by the lack of reliable data on its developmental and (sub)cellular expression patterns. We provide here the first properly controlled analysis of NKCC1 protein expression in various cell types of the mouse brain using custom-made antibodies and an NKCC1 knock-out validated immunohistochemical procedure, with parallel data based on advanced mRNA approaches. NKCC1 protein and mRNA are expressed at remarkably high levels in oligodendrocytes. In immature neurons, NKCC1 protein was located in the somata, whereas in adult neurons, only NKCC1 mRNA could be clearly detected. NKCC1 immunoreactivity is also seen in microglia, astrocytes, developing pericytes, and in progenitor cells of the dentate gyrus. Finally, a differential expression of NKCC1 splice variants was observed, with NKCC1a predominating in non-neuronal cells and NKCC1b in neurons. Taken together, our data provide a cellular basis for understanding NKCC1 functions in the brain and enable the identification of major limitations and promises in the development of neuron-targeting NKCC1-blockers.
Collapse
Affiliation(s)
- Samu N Kurki
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Pavel Uvarov
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Alexey S Pospelov
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Kalevi Trontti
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
- University of Helsinki SleepWell Research Program, Faculty of Medicine, , 00014 Helsinki , Finland
- University of Helsinki Department of Psychology and Logopedics, , 00014 Helsinki , Finland
| | - Antje K Hübner
- Jena University Hospital, Friedrich Schiller Universität Institute of Human Genetics, , 07747 Jena , Germany
| | - Rakenduvadhana Srinivasan
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Masahiko Watanabe
- Hokkaido University Department of Anatomy, Faculty of Medicine, , Sapporo 060–8638 , Japan
| | - Iiris Hovatta
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
- University of Helsinki SleepWell Research Program, Faculty of Medicine, , 00014 Helsinki , Finland
- University of Helsinki Department of Psychology and Logopedics, , 00014 Helsinki , Finland
| | - Christian A Hübner
- Jena University Hospital, Friedrich Schiller Universität Institute of Human Genetics, , 07747 Jena , Germany
| | - Kai Kaila
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Mari A Virtanen
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| |
Collapse
|
9
|
Auer F, Schoppik D. The Larval Zebrafish Vestibular System Is a Promising Model to Understand the Role of Myelin in Neural Circuits. Front Neurosci 2022; 16:904765. [PMID: 35600621 PMCID: PMC9122096 DOI: 10.3389/fnins.2022.904765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/11/2022] [Indexed: 12/27/2022] Open
Abstract
Myelin is classically known for its role in facilitating nerve conduction. However, recent work casts myelin as a key player in both proper neuronal circuit development and function. With this expanding role comes a demand for new approaches to characterize and perturb myelin in the context of tractable neural circuits as they mature. Here we argue that the simplicity, strong conservation, and clinical relevance of the vestibular system offer a way forward. Further, the tractability of the larval zebrafish affords a uniquely powerful means to test open hypotheses of myelin's role in normal development and disordered vestibular circuits. We end by identifying key open questions in myelin neurobiology that the zebrafish vestibular system is particularly well-suited to address.
Collapse
Affiliation(s)
| | - David Schoppik
- Departments of Otolaryngology, Neuroscience & Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
10
|
Activation of the unfolded protein response by Connexin47 mutations associated with Pelizaeus-Merzbacher-like disease. Mol Cell Neurosci 2022; 120:103716. [PMID: 35276347 DOI: 10.1016/j.mcn.2022.103716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 11/23/2022] Open
Abstract
Pelizaeus-Merzbacher-like disease type 1 (PMLD1) is a hypomyelinating disorder arising in patients with mutations in GJC2, encoding Connexin47 (Cx47). PMLD1 causes nystagmus, cerebellar ataxia, spasticity and changes in CNS white matter detected by MRI. At least one mutation (p.I33M) yields a much milder phenotype, spastic paraplegia type 44 (SPG44). Cx47 contributes to gap junction communication channels between oligodendrocytes (OLs), the myelinating cells in the central nervous system (CNS), and between OLs and astrocytes. Prior studies in cell lines have shown that PMLD1 mutants such as p.P87S display defective protein trafficking, intracellular retention in the ER and loss-of-function. Here we show that when expressed in primary OLs, three PMLD1 associated mutants (p.P87S, p.Y269D and p.M283T) show ER retention of Cx47 and evidence of activation of the cellular stress (unfolded protein response, UPR) and apoptotic pathways. On the other hand, the milder SPG44 associated mutation p.I33M shows a wild-type-like subcellular distribution and no activation of the UPR or apoptotic pathways. These studies provide new insight into a potential element of toxic gain of function underlying the mechanism of PMLD1 that should help guide future therapeutic approaches.
Collapse
|
11
|
Loss of monocarboxylate transporter 1 aggravates white matter injury after experimental subarachnoid hemorrhage in rats. Front Med 2021; 15:887-902. [PMID: 34874512 DOI: 10.1007/s11684-021-0879-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/25/2021] [Indexed: 11/26/2022]
Abstract
Monocarboxylic acid transporter 1 (MCT1) maintains axonal function by transferring lactic acid from oligodendrocytes to axons. Subarachnoid hemorrhage (SAH) induces white matter injury, but the involvement of MCT1 is unclear. In this study, the SAH model of adult male Sprague-Dawley rats was used to explore the role of MCT1 in white matter injury after SAH. At 48 h after SAH, oligodendrocyte MCT1 was significantly reduced, and the exogenous overexpression of MCT1 significantly improved white matter integrity and long-term cognitive function. Motor training after SAH significantly increased the number of ITPR2+SOX10+ oligodendrocytes and upregulated the level of MCT1, which was positively correlated with the behavioral ability of rats. In addition, miR-29b and miR-124 levels were significantly increased in SAH rats compared with non-SAH rats. Further intervention experiments showed that miR-29b and miR-124 could negatively regulate the level of MCT1. This study confirmed that the loss of MCT1 may be one of the mechanisms of white matter damage after SAH and may be caused by the negative regulation of miR-29b and miR-124. MCT1 may be involved in the neurological improvement of rehabilitation training after SAH.
Collapse
|
12
|
Kamen Y, Pivonkova H, Evans KA, Káradóttir RT. A Matter of State: Diversity in Oligodendrocyte Lineage Cells. Neuroscientist 2021; 28:144-162. [PMID: 33567971 DOI: 10.1177/1073858420987208] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) give rise to oligodendrocytes which myelinate axons in the central nervous system. Although classically thought to be a homogeneous population, OPCs are reported to have different developmental origins and display regional and temporal diversity in their transcriptome, response to growth factors, and physiological properties. Similarly, evidence is accumulating that myelinating oligodendrocytes display transcriptional heterogeneity. Analyzing this reported heterogeneity suggests that OPCs, and perhaps also myelinating oligodendrocytes, may exist in different functional cell states. Here, we review the evidence indicating that OPCs and oligodendrocytes are diverse, and we discuss the implications of functional OPC states for myelination in the adult brain and for myelin repair.
Collapse
Affiliation(s)
- Yasmine Kamen
- Wellcome-Medical Research Council Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Helena Pivonkova
- Wellcome-Medical Research Council Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Kimberley A Evans
- Wellcome-Medical Research Council Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Ragnhildur T Káradóttir
- Wellcome-Medical Research Council Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, UK.,Department of Physiology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
13
|
Spitzer SO, Sitnikov S, Kamen Y, Evans KA, Kronenberg-Versteeg D, Dietmann S, de Faria O, Agathou S, Káradóttir RT. Oligodendrocyte Progenitor Cells Become Regionally Diverse and Heterogeneous with Age. Neuron 2019; 101:459-471.e5. [PMID: 30654924 PMCID: PMC6372724 DOI: 10.1016/j.neuron.2018.12.020] [Citation(s) in RCA: 246] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/12/2018] [Accepted: 12/12/2018] [Indexed: 01/12/2023]
Abstract
Oligodendrocyte progenitor cells (OPCs), which differentiate into myelinating oligodendrocytes during CNS development, are the main proliferative cells in the adult brain. OPCs are conventionally considered a homogeneous population, particularly with respect to their electrophysiological properties, but this has been debated. We show, by using single-cell electrophysiological recordings, that OPCs start out as a homogeneous population but become functionally heterogeneous, varying both within and between brain regions and with age. These electrophysiological changes in OPCs correlate with the differentiation potential of OPCs; thus, they may underlie the differentiational differences in OPCs between regions and, likewise, differentiation failure with age. Oligodendrocyte progenitor cells (OPCs) acquire ion channels with age OPCs become functionally heterogeneous both between brain regions and with age NMDARs disappear in non-myelinating regions but remain in actively myelinating areas Heterogeneity in OPC ion channel density indicates different functional states
Collapse
Affiliation(s)
- Sonia Olivia Spitzer
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Sergey Sitnikov
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Yasmine Kamen
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Kimberley Anne Evans
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Deborah Kronenberg-Versteeg
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Sabine Dietmann
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Omar de Faria
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Sylvia Agathou
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Ragnhildur Thóra Káradóttir
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
14
|
Marques S, Zeisel A, Codeluppi S, van Bruggen D, Mendanha Falcão A, Xiao L, Li H, Häring M, Hochgerner H, Romanov RA, Gyllborg D, Muñoz Manchado A, La Manno G, Lönnerberg P, Floriddia EM, Rezayee F, Ernfors P, Arenas E, Hjerling-Leffler J, Harkany T, Richardson WD, Linnarsson S, Castelo-Branco G. Oligodendrocyte heterogeneity in the mouse juvenile and adult central nervous system. Science 2016; 352:1326-1329. [PMID: 27284195 PMCID: PMC5221728 DOI: 10.1126/science.aaf6463] [Citation(s) in RCA: 753] [Impact Index Per Article: 83.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/10/2016] [Indexed: 12/15/2022]
Abstract
Oligodendrocytes have been considered as a functionally homogeneous population in the central nervous system (CNS). We performed single-cell RNA sequencing on 5072 cells of the oligodendrocyte lineage from 10 regions of the mouse juvenile and adult CNS. Thirteen distinct populations were identified, 12 of which represent a continuum from Pdgfra(+) oligodendrocyte precursor cells (OPCs) to distinct mature oligodendrocytes. Initial stages of differentiation were similar across the juvenile CNS, whereas subsets of mature oligodendrocytes were enriched in specific regions in the adult brain. Newly formed oligodendrocytes were detected in the adult CNS and were responsive to complex motor learning. A second Pdgfra(+) population, distinct from OPCs, was found along vessels. Our study reveals the dynamics of oligodendrocyte differentiation and maturation, uncoupling them at a transcriptional level and highlighting oligodendrocyte heterogeneity in the CNS.
Collapse
Affiliation(s)
- Sueli Marques
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Amit Zeisel
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Simone Codeluppi
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet SE-17177 Stockholm, Sweden
| | - David van Bruggen
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Ana Mendanha Falcão
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Lin Xiao
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, United Kingdom
- Institute of Neuroscience, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Huiliang Li
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Martin Häring
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Hannah Hochgerner
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Roman A. Romanov
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Austria
| | - Daniel Gyllborg
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Ana Muñoz Manchado
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Gioele La Manno
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Peter Lönnerberg
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Elisa M. Floriddia
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Fatemah Rezayee
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Patrik Ernfors
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Ernest Arenas
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Jens Hjerling-Leffler
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Tibor Harkany
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Austria
| | - William D. Richardson
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Sten Linnarsson
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17177 Stockholm, Sweden
| |
Collapse
|
15
|
Gautier HOB, Evans KA, Volbracht K, James R, Sitnikov S, Lundgaard I, James F, Lao-Peregrin C, Reynolds R, Franklin RJM, Káradóttir RT. Neuronal activity regulates remyelination via glutamate signalling to oligodendrocyte progenitors. Nat Commun 2015; 6:8518. [PMID: 26439639 PMCID: PMC4600759 DOI: 10.1038/ncomms9518] [Citation(s) in RCA: 205] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 08/30/2015] [Indexed: 12/20/2022] Open
Abstract
Myelin regeneration can occur spontaneously in demyelinating diseases such as multiple sclerosis (MS). However, the underlying mechanisms and causes of its frequent failure remain incompletely understood. Here we show, using an in-vivo remyelination model, that demyelinated axons are electrically active and generate de novo synapses with recruited oligodendrocyte progenitor cells (OPCs), which, early after lesion induction, sense neuronal activity by expressing AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid)/kainate receptors. Blocking neuronal activity, axonal vesicular release or AMPA receptors in demyelinated lesions results in reduced remyelination. In the absence of neuronal activity there is a ∼6-fold increase in OPC number within the lesions and a reduced proportion of differentiated oligodendrocytes. These findings reveal that neuronal activity and release of glutamate instruct OPCs to differentiate into new myelinating oligodendrocytes that recover lost function. Co-localization of OPCs with the presynaptic protein VGluT2 in MS lesions implies that this mechanism may provide novel targets to therapeutically enhance remyelination.
Collapse
Affiliation(s)
- Hélène O. B. Gautier
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Kimberley A. Evans
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Katrin Volbracht
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Rachel James
- Faculty of Medicine, Division of Brain Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Sergey Sitnikov
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Iben Lundgaard
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Fiona James
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Cristina Lao-Peregrin
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Richard Reynolds
- Faculty of Medicine, Division of Brain Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Robin J. M. Franklin
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0AH, UK
| | - Ragnhildur T Káradóttir
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| |
Collapse
|
16
|
Progressive disorganization of paranodal junctions and compact myelin due to loss of DCC expression by oligodendrocytes. J Neurosci 2014; 34:9768-78. [PMID: 25031414 DOI: 10.1523/jneurosci.0448-14.2014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Paranodal axoglial junctions are critical for maintaining the segregation of axonal domains along myelinated axons; however, the proteins required to organize and maintain this structure are not fully understood. Netrin-1 and its receptor Deleted in Colorectal Cancer (DCC) are proteins enriched at paranodes that are expressed by neurons and oligodendrocytes. To identify the specific function of DCC expressed by oligodendrocytes in vivo, we selectively eliminated DCC from mature myelinating oligodendrocytes using an inducible cre regulated by the proteolipid protein promoter. We demonstrate that DCC deletion results in progressive disruption of the organization of axonal domains, myelin ultrastructure, and myelin protein composition. Conditional DCC knock-out mice develop balance and coordination deficits and exhibit decreased conduction velocity. We conclude that DCC expression by oligodendrocytes is required for the maintenance and stability of myelin in vivo, which is essential for proper signal conduction in the CNS.
Collapse
|
17
|
Miller DJ, Lackey EP, Hackett TA, Kaas JH. Development of myelination and cholinergic innervation in the central auditory system of a prosimian primate (Otolemur garnetti). J Comp Neurol 2014; 521:3804-16. [PMID: 23749337 DOI: 10.1002/cne.23379] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 05/22/2013] [Accepted: 05/23/2013] [Indexed: 11/07/2022]
Abstract
Change in the timeline of neurobiological growth is an important source of biological variation, and thus phenotypic evolution. However, no study has to date investigated sensory system development in any of the prosimian primates that are thought to most closely resemble our earliest primate ancestors. Acetylcholine (ACh) is a neurotransmitter critical to normal brain function by regulating synaptic plasticity associated with attention and learning. Myelination is an important structural component of the brain because it facilitates rapid neuronal communication. In this work we investigated the expression of acetylcholinesterase (AChE) and the density of myelinated axons throughout postnatal development in the inferior colliculus (IC), medial geniculate complex (MGC), and auditory cortex (auditory core, belt, and parabelt) in Garnett's greater galago (Otolemur garnetti). We found that the IC and MGC exhibit relatively high myelinated fiber length density (MFLD) values at birth and attain adult-like values by the species-typical age at weaning. In contrast, neocortical auditory fields are relatively unmyelinated at birth and only attain adult-like MFLD values by the species-typical age at puberty. Analysis of AChE expression indicated that, in contrast to evidence from rodent samples, the adult-like distribution of AChE in the core area of auditory cortex, dense bands in layers I, IIIb/IV, and Vb/VI, is present at birth. These data indicate the differential developmental trajectory of central auditory system structures and demonstrate the early onset of adult-like AChE expression in primary auditory cortex in O. garnetti, suggesting the auditory system is more developed at birth in primates compared to rodents.
Collapse
Affiliation(s)
- Daniel J Miller
- Department of Psychology, Vanderbilt University, Nashville, Tennessee, 37205
| | | | | | | |
Collapse
|
18
|
Koshiya N, Oku Y, Yokota S, Oyamada Y, Yasui Y, Okada Y. Anatomical and functional pathways of rhythmogenic inspiratory premotor information flow originating in the pre-Bötzinger complex in the rat medulla. Neuroscience 2014; 268:194-211. [DOI: 10.1016/j.neuroscience.2014.03.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 03/02/2014] [Accepted: 03/04/2014] [Indexed: 01/30/2023]
|
19
|
Dugas JC, Emery B. Purification and culture of oligodendrocyte lineage cells. Cold Spring Harb Protoc 2013; 2013:810-4. [PMID: 24003197 DOI: 10.1101/pdb.top074898] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Oligodendrocytes are the cells of the vertebrate central nervous system responsible for forming myelin sheaths, which are essential for the rapid propagation of action potential. The formation of oligodendrocytes and myelin sheaths is tightly regulated, both temporally and spatially, by a number of extracellular and intracellular factors. For example, notch ligands, thyroid hormones, and mitogens such as platelet-derived growth factor (PDGF) and fibroblast growth factor (FGF) can all interact with oligodendrocyte precursor cell-expressed receptors to impact proliferation, differentiation, and myelin gene expression. To facilitate oligodendrocyte biology research, we have developed a technique using immunopanning to isolate different stages of the oligodendrocyte lineage, oligodendrocyte precursor cells and/or postmitotic oligodendrocytes, from postnatal rat or mouse brains. These cells can be cultured in defined, serum-free media in conditions that either promote differentiation into mature oligodendrocytes or continued proliferation as immature oligodendrocyte precursors. These cells represent an ideal system in which to study the regulation of oligodendrocyte proliferation, migration, differentiation, myelin gene expression, or other fundamental aspects of oligodendrocyte biology.
Collapse
Affiliation(s)
- Jason C Dugas
- Stanford University School of Medicine, Department of Neurobiology, Stanford, California 94305-5125, USA.
| | | |
Collapse
|
20
|
Sherrill LK, Stanis JJ, Gulley JM. Age-dependent effects of repeated amphetamine exposure on working memory in rats. Behav Brain Res 2013; 242:84-94. [PMID: 23291159 DOI: 10.1016/j.bbr.2012.12.044] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 12/20/2012] [Accepted: 12/24/2012] [Indexed: 11/27/2022]
Abstract
Cognitive dysfunction is a hallmark of chronic psychostimulant misuse. Adolescents may have heightened risk of developing drug-induced deficits because their brains are already undergoing widespread changes in anatomy and function as a normal part of development. To address this hypothesis, we performed two sets of experiments where adolescent and young adult rats were pre-exposed to saline or amphetamine (1 or 3mg/kg) and subsequently tested in a prefrontal cortex (PFC)-sensitive working memory task. A total of ten injections of AMPH or saline (in control rats) were given every other day over the course of 19 days. After rats reached adulthood (>90 days old), cognitive performance was assessed using operant-based delayed matching-to-position (DMTP) and delayed nonmatching-to-position (DNMTP) tasks. DNMTP was also assessed following challenges with amphetamine (0.3-1.25mg/kg), and ketamine (5.0-10mg/kg). In experiment one, we also measured the locomotor response following the first and tenth pre-exposure to amphetamine and after an amphetamine challenge given at the conclusion of operant testing. Compared to adult-exposed groups, adolescents were less sensitive to the psychomotor effects of amphetamine. However, they were more vulnerable to exposure-induced cognitive impairments. For example, adolescent-exposed rats displayed delay-dependent deficits in accuracy, increased sensitivity to proactive interference, and required more training to reach criterion. Drug challenges produced deficits in DNMTP performance, but these were not dependent on pre-exposure group. These studies demonstrate age of exposure-dependent effects of amphetamine on cognition in a PFC-sensitive task, suggesting a heightened sensitivity of adolescents to amphetamine-induced neuroplasticity.
Collapse
Affiliation(s)
- Luke K Sherrill
- Department of Psychology, University of Illinois, Urbana-Champaign, USA
| | | | | |
Collapse
|
21
|
Developmental changes and subcellular location in inhibitor of DNA binding 2 (Id2) immunoreactivity in the rat Corpus callosum. Acta Histochem 2012; 114:653-8. [PMID: 22172709 DOI: 10.1016/j.acthis.2011.11.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 11/19/2011] [Accepted: 11/21/2011] [Indexed: 01/21/2023]
Abstract
The mechanisms underlying oligodendrocyte differentiation and myelination are still unclear, but understanding them will be critical for the development of therapies for multiple sclerosis. Inhibitor of DNA binding 2 (Id2) is a transcription factor thought to inhibit oligodendrocyte differentiation, however, it is not known whether the developmental changes and subcellular localization of Id2 are related to myelination. Therefore, we investigated the developmental changes in and the subcellular localization of Id2 immunoreactivity in the rat Corpus callosum, at post-natal developmental stages P0, P7, P14, P21, P42 and P90, by immunohistochemistry. Id2 expression increased from P0 to a peak at P42, the late stage of myelination in the Corpus callosum. Id2 immunostaining decreased slightly, but still remained high at P90. Subcellular localization of Id2 changed from presence in cytoplasm at P14 to the nuclei at P42. Moreover, Id2 was mainly co-localized with CC-1-immunopositive mature oligodendrocytes at P42. These results may be consistent with Id2 inhibitory function in oligodendrocyte differentiation, at the end of myelination or in compaction of myelin in the Corpus callosum of postnatal rat brain.
Collapse
|
22
|
Chen H, Epelbaum S, Delatour B. Fiber Tracts Anomalies in APPxPS1 Transgenic Mice Modeling Alzheimer's Disease. J Aging Res 2011; 2011:281274. [PMID: 21912744 PMCID: PMC3170810 DOI: 10.4061/2011/281274] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2011] [Revised: 05/30/2011] [Accepted: 06/28/2011] [Indexed: 11/20/2022] Open
Abstract
Amyloid beta (Aβ) peptides are known to accumulate in the brain of patients with Alzheimer's disease (AD). However, the link between brain amyloidosis and clinical symptoms has not been elucidated and could be mediated by secondary neuropathological alterations such as fiber tracts anomalies. In the present study, we have investigated the impact of Aβ overproduction in APPxPS1 transgenic mice on the integrity of forebrain axonal bundles (corpus callosum and anterior commissure). We found evidence of fiber tract volume reductions in APPxPS1 mice that were associated with an accelerated age-related loss of axonal neurofilaments and a myelin breakdown. The severity of these defects was neither correlated with the density of amyloid plaques nor associated with cell neurodegeneration. Our data suggest that commissural fiber tract alterations are present in Aβ-overproducing transgenic mice and that intracellular Aβ accumulation preceding extracellular deposits may act as a trigger of such morphological anomalies.
Collapse
Affiliation(s)
- H Chen
- CNRS, Laboratoire NAMC, UMR 8620, Université Paris-Sud 11, 91405 Orsay, France
| | | | | |
Collapse
|
23
|
Umahara T, Uchihara T, Nakamura A, Iwamoto T. Differential expression of 14-3-3 protein isoforms in developing rat hippocampus, cortex, rostral migratory stream, olfactory bulb, and white matter. Brain Res 2011; 1410:1-11. [PMID: 21813115 DOI: 10.1016/j.brainres.2011.06.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Revised: 05/27/2011] [Accepted: 06/15/2011] [Indexed: 11/24/2022]
Abstract
We investigated the differential immunoexpression of 14-3-3 proteins according to their 7 isoforms during the postnatal development of rat brains, primarily in the hippocampus, cortex, rostral migratory stream (RMS), olfactory bulb, and white matter. Wistar rats at different developmental stages, on postnatal days 2 (P2), P7, P14, P21 and P100 were obtained, and were incubated with each type of anti-14-3-3 isoform antibody. 14-3-3 common (COM)-like immunoreactivity (IR) which represents an epitope shared among the 7 isoforms was initially expressed in the olfactory bulb on P2. This IR was partially expressed in the dentate granule cells and hippocampal pyramidal neurons from P7, and increased during development. These chronological changes were similar to those obtained with beta, gamma, and eta isoforms. Epsilon isoform-like IR was initially identified in the cell body of cortical neurons and glia-like cells on P2. After P7, the IR was more intense in the neuropil of the cortex. This epsilon isoform-like IR was markedly accentuated in the stratum lucidum of the hippocampus after P7, where hippocampal mossy fibers terminate, functioning as a giant synapse. This suggests that epsilon isoforms may be associated with synaptogenesis of the hippocampal mossy fibers. Sigma isoform-like IR was observed in the nuclei of external plexiform layer cells of the olfactory bulb from P2 to P21, in the nuclei of the hippocampal pyramidal and dentate granule cells after P7 and in the nuclei of RMS cells after P7. Zeta and tau isoform-like IRs were mainly identified in the white matter and in oligodendroglial cells from P7 to P21. Different immunolocalizations of the 7 isoforms suggest that 14-3-3 protein isoforms are individually associated with neuronal development and synaptogenesis during postnatal formation of the rat brain.
Collapse
Affiliation(s)
- Takahiko Umahara
- Department of Geriatric Medicine, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan.
| | | | | | | |
Collapse
|
24
|
The physiology of developmental changes in BOLD functional imaging signals. Dev Cogn Neurosci 2011; 1:199-216. [PMID: 22436508 DOI: 10.1016/j.dcn.2011.04.001] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 04/18/2011] [Accepted: 04/19/2011] [Indexed: 12/14/2022] Open
Abstract
BOLD fMRI (blood oxygenation level dependent functional magnetic resonance imaging) is increasingly used to detect developmental changes of human brain function that are hypothesized to underlie the maturation of cognitive processes. BOLD signals depend on neuronal activity increasing cerebral blood flow, and are reduced by neural oxygen consumption. Thus, developmental changes of BOLD signals may not reflect altered information processing if there are concomitant changes in neurovascular coupling (the mechanism by which neuronal activity increases blood flow) or neural energy use (and hence oxygen consumption). We review how BOLD signals are generated, and explain the signalling pathways which convert neuronal activity into increased blood flow. We then summarize in broad terms the developmental changes that the brain's neural circuitry undergoes during growth from childhood through adolescence to adulthood, and present the changes in neurovascular coupling mechanisms and energy use which occur over the same period. This information provides a framework for assessing whether the BOLD changes observed during human development reflect altered cognitive processing or changes in neurovascular coupling and energy use.
Collapse
|
25
|
Liu C, Li W, Johnson GA, Wu B. High-field (9.4 T) MRI of brain dysmyelination by quantitative mapping of magnetic susceptibility. Neuroimage 2011; 56:930-8. [PMID: 21320606 DOI: 10.1016/j.neuroimage.2011.02.024] [Citation(s) in RCA: 194] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Revised: 02/04/2011] [Accepted: 02/06/2011] [Indexed: 12/11/2022] Open
Abstract
The multilayered myelin sheath wrapping around nerve axons is essential for proper functioning of the central nervous system. Abnormal myelination leads to a wide range of neurological diseases and developmental disorders. Non-invasive imaging of myelin content is of great clinical importance. The present work demonstrated that loss of myelin in the central nervous system of the shiverer mouse results in a dramatic reduction of magnetic susceptibility in white matter axons. The reduction resulted in a near extinction of susceptibility contrast between gray and white matter. Quantitative magnetic susceptibility imaging and diffusion tensor imaging were conducted on a group of control and shiverer mice at 9.4 T. We measured the resonance frequency distribution of the whole brain for each mouse. Magnetic susceptibility maps were computed and compared between the two groups. It was shown that the susceptibility contrast between gray and white matter was reduced by 96% in the shiverer compared to the controls. Diffusion measurements further confirmed intact fiber pathways in the shiverer mice, ruling out the possibility of axonal injury and its potential contribution to the altered susceptibility. As an autosomal recessive mutation, shiverer is characterized by an almost total lack of central nervous system myelin. Our data provide new evidences indicating that myelin is the predominant source of susceptibility differences between deep gray and white matter observed in magnetic resonance imaging. More importantly, the present study suggests that quantitative magnetic susceptibility is a potential endogenous biomarker for myelination.
Collapse
Affiliation(s)
- Chunlei Liu
- Brain Imaging and Analysis Center, School of Medicine, Duke University, Durham, NC 27705, USA.
| | | | | | | |
Collapse
|
26
|
Bakiri Y, Káradóttir R, Cossell L, Attwell D. Morphological and electrical properties of oligodendrocytes in the white matter of the corpus callosum and cerebellum. J Physiol 2011; 589:559-73. [PMID: 21098009 PMCID: PMC3055543 DOI: 10.1113/jphysiol.2010.201376] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Accepted: 11/22/2010] [Indexed: 11/08/2022] Open
Abstract
In the central nervous system, electrical signals passing along nerve cells are speeded by cells called oligodendrocytes, which wrap the nerve cells with a fatty layer called myelin. This layer is important for rapid information processing, and is often lost in disease, causing mental or physical impairment in multiple sclerosis, stroke, cerebral palsy and spinal cord injury. The myelin speeds the information flow in two ways, by decreasing the capacitance of the nerve cell and by increasing its membrane resistance, but little is known about the latter aspect of myelin function. By recording electrically from oligodendrocytes and imaging their morphology we characterised the geometry and, for the first time, the resistance of myelin in the brain. This revealed differences between the properties of oligodendrocytes in two brain areas and established that the resistance of myelin is sufficiently high to prevent significant slowing of the nerve electrical signal by current leakage through the myelin.
Collapse
Affiliation(s)
- Yamina Bakiri
- Department of Physiology, University College London, London, UK
| | | | | | | |
Collapse
|
27
|
Differential short-term plasticity at convergent inhibitory synapses to the substantia nigra pars reticulata. J Neurosci 2010; 30:14854-61. [PMID: 21048144 DOI: 10.1523/jneurosci.3895-10.2010] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Inhibitory projections from the striatum and globus pallidus converge onto GABAergic projection neurons of the substantia nigra pars reticulata (SNr). Based on existing structural and functional evidence, these pathways are likely to differentially regulate the firing of SNr neurons. We sought to investigate the functional differences in inhibitory striatonigral and pallidonigral traffic using whole-cell voltage clamp in brain slices with these pathways preserved. We found that striatonigral IPSCs exhibited a high degree of paired-pulse facilitation. We tracked this facilitation over development and found the facilitation as the animal aged, but stabilized by postnatal day 17 (P17), with a paired pulse ratio of 2. We also found that the recovery from facilitation accelerated over development, again, reaching a stable phenotype by P17. In contrast, pallidonigral synapses show paired-pulse depression, and this depression could be solely explained by presynaptic changes. The mean paired-pulse ratio of 0.67 did not change over development, but the recovery from depression slowed over development. Pallidonigral IPSCs were significantly faster than striatonigral IPSCs when measured at the soma. Finally, under current clamp, prolonged bursts of striatal IPSPs were able to consistently silence the pacemaker activity of nigral neurons, whereas pallidal inputs depressed, allowing nigral neurons to reinstate firing. These findings highlight the importance of differential dynamics of neurotransmitter release in regulating the circuit behavior of the basal ganglia.
Collapse
|
28
|
Yuan W, Deren KE, McAllister JP, Holland SK, Lindquist DM, Cancelliere A, Mason M, Shereen A, Hertzler DA, Altaye M, Mangano FT. Diffusion tensor imaging correlates with cytopathology in a rat model of neonatal hydrocephalus. Cerebrospinal Fluid Res 2010; 7:19. [PMID: 21054844 PMCID: PMC2989304 DOI: 10.1186/1743-8454-7-19] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Accepted: 11/05/2010] [Indexed: 11/23/2022] Open
Abstract
Background Diffusion tensor imaging (DTI) is a non-invasive MRI technique that has been used to quantify CNS abnormalities in various pathologic conditions. This study was designed to quantify the anisotropic diffusion properties in the brain of neonatal rats with hydrocephalus (HCP) and to investigate association between DTI measurements and cytopathology. Methods DTI data were acquired between postnatal day 7 (P7) and P12 in 12 rats with HCP induced at P2 and in 15 age-matched controls. Animals were euthanized at P11 or P22/P23 and brains were processed with immunohistochemistry for glial fibrillary acidic protein (GFAP), ionized calcium-binding adaptor molecule (Iba-1), and luxol fast blue (LFB) to assess astrocytosis, microglial reactivity and degree of myelination, respectively. Results Hydrocephalic rats were consistently found to have an abnormally low (at corrected p-level of <0.05) fractional anisotropy (FA) value and an abnormally high mean diffusivity (MD) value in the cerebral cortex (CX), the corpus callosum (CC), and the internal capsule (IC). Immunohistochemical analysis demonstrated trends of increasing astrocyte and microglial reactivity in HCP rats at P11 that reached statistical significance at P22/P23. A trend toward reduced myelination in the HCP rats was also found at P22/P23. Correlation analysis at P11 for the CC demonstrated statistically significant correlations (or trends) between the DTI measurement (the decreased FA and increased MD values) and the GFAP or Iba-1 rankings. The immunohistochemical rankings in the IC at P22/P23 were also significantly correlated or demonstrated a trend with both FA and MD values. Conclusions This study demonstrates the feasibility of employing DTI on the brain in experimental hydrocephalus in neonatal rats and reveals impairments in multiple regions of interest in both grey and white matter. A strong correlation was found between the immunohistochemical results and the changes in anisotropic diffusion properties.
Collapse
Affiliation(s)
- Weihong Yuan
- Division of Pediatric Neurosurgery, University of Cincinnati, Cincinnati Children's Hospital Medical Center MLC 2016, 3333 Burnet Avenue, Cincinnati, OH 45229, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Is activity regulation of late myelination a plastic mechanism in the human nervous system? ACTA ACUST UNITED AC 2009; 5:29-34. [PMID: 19785923 DOI: 10.1017/s1740925x09990330] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Studies on various animal models have established that neuronal activity can influence the myelination process. Are such mechanisms present in humans, and do they mediate experience-driven white matter plasticity not only during early development but also in adolescents and adults? While there is as yet no direct evidence for this, a number of findings - reviewed here - are consistent with this idea. First, postmortem and neuroimaging studies show that the human white matter development is a protracted process that continues well into adulthood. Second, developmental changes and individual differences in white matter structure are related to differences in neural activity and behavior. Finally, studies on effects of long-term training, in particular in musicians, show strong relations between training and white matter structure. I conclude by briefly discussing possible types of white matter plasticity that could underlie these findings, emphasizing a distinction between indirect myelination plasticity, where the myelin sheath grows in parallel with the axon itself, and direct myelination plasticity, where the myelin sheath thickness is modulated independently of axonal diameter.
Collapse
|
30
|
Wang S, Wu EX, Cai K, Lau HF, Cheung PT, Khong PL. Mild hypoxic-ischemic injury in the neonatal rat brain: longitudinal evaluation of white matter using diffusion tensor MR imaging. AJNR Am J Neuroradiol 2009; 30:1907-13. [PMID: 19749219 DOI: 10.3174/ajnr.a1697] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND PURPOSE Selective white matter (WM) damage is a known sequela of mild hypoxic-ischemic (HI) injury in the neonatal rat model. The aim of this study was to evaluate longitudinally mild HI-induced WM damage (represented by the external capsule [EC]) by diffusion tensor MR imaging (DTI) and to correlate the findings with histology. MATERIALS AND METHODS Seven-day-old Sprague-Dawley rats (n = 19) underwent unilateral ligation of the left common carotid artery followed by hypoxia for 50 minutes to create mild HI injury. DTI was performed longitudinally at 5 time points from day 1 to day 90 postinjury (n = 19, 16, 13, 11, 9, respectively), and fractional anisotropy (FA), trace, radial diffusivity (lambda( perpendicular)), and axial diffusivity (lambda(//)) of the injury and control contralateral ECs were quantified. Rats were randomly sacrificed (n = 15, in total), and the corresponding ECs were stained with hematoxylin-eosin, Luxol fast blue (LFB), and neurofilament (NF) to evaluate morphologic changes, amount of myelin, and axonal count at every time point. A paired t test was applied to evaluate statistical differences between both ECs, and the Pearson correlation test was used to evaluate the relationships between DTI indices and histologic evaluations. In addition, longitudinal changes in DTI indices and histologic evaluations were analyzed by a linear mixed model and an analysis of variance test, respectively. RESULTS We demonstrated significantly decreased FA, increased lambda( perpendicular), and similar lambda(//) in the injury compared with the control EC, which was persistent through all time points. Histologic evaluation by LFB and NF staining showed reduced myelin stain intensity in the injury EC and similar axonal counts in both ECs. Longitudinally, there was an increase in FA, a decrease in lambda( perpendicular) and trace, and stability in lambda(//) in both ECs. Also, there was progressive reduction in the differences in FA, trace, and lambda( perpendicular) between the injury and control EC, especially between day 1 and day 7 postinjury and in tandem with changes in myelin stain. FA was significantly correlated with myelin stain (r = 0.681, P < .01) and axonal count (r = 0.673, P < .01), whereas lambda( perpendicular) was significantly correlated with myelin stain only (r = -0.528, P < .01), and lambda(//), with axonal count only (r = 0.372, P = .043). CONCLUSIONS Diffusion indices can reflect dysmyelination in mild HI injury, continual myelination of both injury and control ECs with growth, and the partial recovery of myelin postinjury. We propose that diffusion indices may be used as biomarkers to monitor noninvasively the longitudinal changes of mild HI-induced WM damage.
Collapse
Affiliation(s)
- S Wang
- Department of Diagnostic Radiology, University of Hong Kong, Hong Kong, People's Republic of China
| | | | | | | | | | | |
Collapse
|
31
|
Keller AF, Gravel M, Kriz J. Live imaging of amyotrophic lateral sclerosis pathogenesis: disease onset is characterized by marked induction of GFAP in Schwann cells. Glia 2009; 57:1130-42. [PMID: 19115383 DOI: 10.1002/glia.20836] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a late-onset neurological disease characterized by progressive loss of motor neurons. At present, the pathological events precipitating disease onset and the exact pattern of disease progression are not fully understood. Recent studies suggest that glial cells, in particular activated astrocytes, can release factors that can directly kill motor neurons. To further investigate the involvement of glial cells (astrocytes and Schwann cells) in the pathogenesis of ALS, we generated ALS-(GFAP-luciferase/SOD(G93A)) reporter mouse in which upregulation of glial fibrillary acidic protein (GFAP) can be visualized from live animals throughout the different stages of disease. Our results suggest that the disease in mice is initiated simultaneously in the spinal cord and in the peripheral nerves and is characterized by several cycles of GFAP upregulation. Immunohistochemical analysis confirmed that the induction GFAP bioluminescence signals were associated with the significant increases in GFAP immunoreactivity. The first pathological GFAP signals occurring at 25-30 days were asymptomatic and detectable at the level of lumbar spinal cord projections and at the periphery. These early events were then followed by GFAP promoter inductions that were associated with the distinct clinical symptoms. As expected, the onset of paralysis (112 days) was associated with the gradual and marked GFAP upregulation in the spinal cord. Interestingly, however, the disease onset (90 days) was characterized by sharp and synchronized induction of GFAP in peripheral nerve Schwann cells suggesting that peripheral nerves pathology/denervation and associated Schwann cell stress may play an important role in the ALS pathogenesis.
Collapse
Affiliation(s)
- A Florence Keller
- Department of Anatomy and Physiology, Laval University, Centre de Recherche du Centre Hospitalier de l'Université Laval, Quebec City, Quebec G1V 4G2, Canada
| | | | | |
Collapse
|
32
|
Yang HJ, Wang H, Zhang Y, Xiao L, Clough RW, Browning R, Li XM, Xu H. Region-specific susceptibilities to cuprizone-induced lesions in the mouse forebrain: Implications for the pathophysiology of schizophrenia. Brain Res 2009; 1270:121-30. [PMID: 19306847 DOI: 10.1016/j.brainres.2009.03.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2009] [Revised: 03/03/2009] [Accepted: 03/05/2009] [Indexed: 01/13/2023]
Abstract
Cuprizone (CPZ) is a neurotoxic agent acting as a copper chelator. In our recent study, C57BL/6 mice given dietary CPZ (0.2%) showed impairments in spatial working memory, social interaction, and prepulse inhibition. These abnormalities are reminiscent of certain schizophrenia symptoms and are not likely due to damage in the whole brain or in any single white matter tract/brain region. We hypothesized that white matter damage resulting from CPZ-treatment may be site-specific rather than universal. We examined the forebrains of C57BL/6 mice given the CPZ-containing diet and compared them with those of controls. We assessed CPZ-induced demyelination in main white matter tracts of the forebrain, evaluated myelin break down in the neuropil of the main olfactory bulb (MOB), cerebral cortex (CTX), caudate putamen (CP), hippocampus (HP), thalamus (TH), and hypothalamus (HY), and counted the number of myelin sheath forming oligodendrocytes (OLs) in CTX, CP, TH, and HY. Obvious demyelination was observed in the corpus callosum, external capsule, CP, and dorsal hippocampal commissure whereas other tracts seemed to be unaffected. The neuropil of CTX, HP and MOB showed myelin break down, which was mild in TH and HY. The number of OLs was decreased in all the above regions of CPZ-treated mice although the degree of OL loss was not consistent across regions. The data provide further support for white matter abnormalities contributing to schizophrenia-like behaviors in mice.
Collapse
Affiliation(s)
- Hong-Ju Yang
- Department of Anatomy, School of Medicine, Southern Illinois University Carbondale, 1135 Lincoln Dr., Carbondale, IL 62901, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
López V, Wagner CK. Progestin receptor is transiently expressed perinatally in neurons of the rat isocortex. J Comp Neurol 2009; 512:124-39. [PMID: 18973223 DOI: 10.1002/cne.21883] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Steroid hormones influence the development of numerous brain regions, including some that are not classically considered steroid-sensitive. For example, nuclear receptors for both androgen and estrogen have been detected in neonatal cortical cells. High levels of progestin binding and progestin receptor (PR) mRNA have also been reported in early perinatal isocortex. PR expression coincides with high levels of de novo progesterone produced within the cortex, suggesting that PR and its ligand influence the important developmental cortical processes occurring shortly after birth. In order to better understand the role PR plays in cortical development, we used the cellular-level resolution of immunohistochemistry and in situ hybridization (ISH) to characterize changes in perinatal PR expression within specific cortical lamina. PR immunoreactivity (PR-ir) was examined at embryonic days (E) 18, 20, 21, 22, and postnatal days (P) 1, 3, 6, 9, 13, and 27. We find that PR-ir is transiently expressed in specific lamina of frontal, parietal, temporal, and occipital cortex. PR-ir was observed in subplate cells on E18, in increasingly superficial lamina (primarily lamina V, then II/III) during early postnatal development, and was absent by P27. Double-labeling immunohistochemistry indicated that PR-ir colocalizes with the neuronal marker, microtubule associated protein-2, but not with the glial marker, nestin, nor with gamma-aminobutyric acid. These results suggest that specific subpopulations of cortical neurons may be transiently sensitive to progesterone, and that progesterone and its receptor may play a critical role in the fundamental mechanisms underlying normal cortical development.
Collapse
Affiliation(s)
- Verónica López
- Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003, USA.
| | | |
Collapse
|
34
|
Umahara T, Uchihara T, Nakamura A, Iwamoto T. Isoform-dependent immunolocalization of 14-3-3 proteins in developing rat cerebellum. Brain Res 2008; 1253:15-26. [PMID: 19070608 DOI: 10.1016/j.brainres.2008.11.065] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Accepted: 11/17/2008] [Indexed: 11/26/2022]
Abstract
We investigated the expression of 14-3-3 protein and its 7 isoforms during postnatal development of rat cerebellum with immunoblot and immunohistochemistry with isoform-specific antibodies. The relative amounts of total 14-3-3 protein, probed by an antibody (14-3-3 COM) recognizing a sequence shared among its isoforms, exhibited no significant changes from postnatal day 2 (P2) to P100. 14-3-3 COM-like immunoreactivity (IR), initially in the apical portion of Purkinje cells at P2, extended to Purkinje cell bodies at P14 and to their dendrites (P100) with increasing intensity. Molecular layer (after P7) and cerebellar nucleus neurons (after P14) were also immunolabeled with this antibody. These chronological changes were shared with those obtained with beta, gamma, and eta isoforms. In contrast, epsilon isoform-like IR was initially identified in processes of radial and Bergmann glia at P2 prior to its appearance in the molecular layer at P7 with subsequent intensification also in Purkinje cells after P14. Zeta and tau isoform-like IR was identified in the white matter and/or in oligodendroglial cells. The sigma isoform was the only isoform exhibiting a significant quantitative change with a peak at P14. Immunolocalization of sigma isoform was initially restricted in several cells in Purkinje cell layer at P2 and shifted to nuclei of external and internal granule cells and Purkinje cells after P14, whereas its immunolabeling was markedly weaker at P100. Different immunolocalizations of the 7 isoforms suggest that 14-3-3 protein isoforms individually associate with the neuronal and glial proliferation, differentiation, migration and development during postnatal formation of rat cerebellum.
Collapse
Affiliation(s)
- Takahiko Umahara
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan.
| | | | | | | |
Collapse
|
35
|
Alpeeva EV, Makarenko IG. Perinatal development of the mammillothalamic tract and innervation of the anterior thalamic nuclei. Brain Res 2008; 1248:1-13. [PMID: 19026995 DOI: 10.1016/j.brainres.2008.10.060] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2008] [Revised: 10/21/2008] [Accepted: 10/22/2008] [Indexed: 01/07/2023]
Abstract
Axonal projections originating from the mammillary bodies represent important pathways that are essential for spatial information processing. Mammillothalamic tract is one of the main efferent projection systems of the mammillary body belonging to the limbic "Papez circuit". This study was aimed to describe the schedule of the mammillothalamic tract development in the rat using carbocyanine dye tracing. It was shown for the first time that fibers of the mammillothalamic tract being the collaterals of the mammillotegmental tract axons start bifurcating from the mammillotegmental tract on E17. The axons of the mammillothalamic tract grow simultaneously and reach the ventral region of the anterior thalamus where they form first terminal arborizations on E20-E21. Ipsilateral projections from the medial mammillary nucleus to the anteromedial and anteroventral thalamic nuclei develop from E20 to P6. Bilateral projections from the lateral mammillary nucleus to the anterodorsal thalamic nuclei develop later, on P3-P6, after the formation of the thalamic decussation of the mammillary body axons. Unique spatial and temporal pattern of the perinatal development of ascending mammillary body projections to the anterior thalamic nuclei may reflect the importance of these connections within the limbic circuitry.
Collapse
Affiliation(s)
- E V Alpeeva
- Optical Research Group, Koltsov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov str., 119334 Moscow, Russian Federation
| | | |
Collapse
|
36
|
Fong AY, Corcoran AE, Zimmer MB, Andrade DV, Milsom WK. Respiratory rhythm of brainstem-spinal cord preparations: Effects of maturation, age, mass and oxygenation. Respir Physiol Neurobiol 2008; 164:429-40. [PMID: 18948229 DOI: 10.1016/j.resp.2008.09.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2008] [Revised: 08/28/2008] [Accepted: 09/12/2008] [Indexed: 10/21/2022]
Abstract
We examined the effect of age, mass and the presence of the pons on the longevity (length of time spontaneous respiratory-related activity is produced) of brainstem-spinal cord preparations of neonatal rodents (rats and hamsters) and the level of oxygenation in the medulla respiratory network in these preparations. We found the longevity of the preparations from both species decreased with increasing postnatal age. Physical removal of the pons increased respiratory frequency and the longevity of the preparation. However, tissue oxygenation at the level of the medullary respiratory network was not affected by removal of the pons or increasing postnatal age (up to postnatal day 4). Taken together, these data suggest that the effect of removing the pons on respiratory frequency and the longevity of brainstem-spinal cord preparations with increasing postnatal age are primarily due to postnatal development and appear to be unrelated to mass or changes in levels of tissue oxygenation.
Collapse
Affiliation(s)
- Angelina Y Fong
- Department of Zoology, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada.
| | | | | | | | | |
Collapse
|
37
|
Bockhorst KH, Narayana PA, Liu R, Ahobila-Vijjula P, Ramu J, Kamel M, Wosik J, Bockhorst T, Hahn K, Hasan KM, Perez-Polo JR. Early postnatal development of rat brain: in vivo diffusion tensor imaging. J Neurosci Res 2008; 86:1520-8. [PMID: 18189320 DOI: 10.1002/jnr.21607] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Perinatal hypoxia is a major cause of neurodevelopmental deficits. Neuronal migration patterns are particularly sensitive to perinatal hypoxia/ischemia and are associated with the clinical deficits. The rat model of hypoxia/ischemia at P7 mimics that of perinatal injury in humans. Before assessing the effects of postnatal injury on brain development, it is essential to determine the normal developmental trajectories of various brain structures in individual animals. In vivo longitudinal diffusion tensor imaging (DTI) was performed from postnatal day 0 (P0) to P56 on Wistar rats. The DTI metrics, mean diffusivity (MD), fractional anisotropy (FA), axial (lambdal) and radial (lambdat) diffusivities, were determined for four gray matter and eight white matter structures. The FA of the cortical plate and the body of corpus callosum decreased significantly during the first 3 weeks after birth. The decrease in the cortical plate's FA value was associated mainly with an increase in lambdat. The initial decrease in FA of corpus callosum was associated with a significant decrease in lambdal. The FA of corpus callosum increased during the rest of the observational period, which was mainly associated with a decrease in lambdat. The FA of gray matter structures, hippocampus, caudate putamen, and cortical mantle did not show significant changes between P0 and P56. In contrast, the majority of white matter structures showed significant changes between P0 and P56. These temporal changes in the DTI metrics were related to the neuronal and axonal pruning and myelination that are known to occur in the developing brain.
Collapse
Affiliation(s)
- K H Bockhorst
- University of Texas at Houston, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Oligodendrocytes (OGs) assemble the myelin sheath around axons in the central nervous system. Specification of cells into the OG lineage is largely the result of interplay between bone morphogenetic protein, sonic hedgehog and Notch signaling pathways, which regulate expression of transcription factors (TFs) dictating spatial and temporal aspects of oligodendrogenesis. Many of these TFs and others then direct OG development through to a mature myelinating OG. Here we describe signaling pathways and TFs that are inductive, inhibitory, and/or permissive to OG specification and maturation. We develop a basic transcriptional network and identify similarities and differences between regulation of oligodendrogenesis in the spinal cord and brain.
Collapse
Affiliation(s)
- Danette J Nicolay
- Laboratory of Molecular Biology, College of Pharmacy and Nutrition, University of Saskatchewan, and Cameco MS Neuroscience Research Center, City Hospital, Saskatoon, Saskatchewan, Canada
| | | | | |
Collapse
|
39
|
Shoykhet M, Simons DJ. Development of thalamocortical response transformations in the rat whisker-barrel system. J Neurophysiol 2007; 99:356-66. [PMID: 17989240 DOI: 10.1152/jn.01063.2007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Extracellular single-unit recordings were used to characterize responses of thalamic barreloid and cortical barrel neurons to controlled whisker deflections in 2, 3-, and 4-wk-old and adult rats in vivo under fentanyl analgesia. Results indicate that response properties of thalamic and cortical neurons diverge during development. Responses to deflection onsets and offsets among thalamic neurons mature in parallel, whereas among cortical neurons responses to deflection offsets become disproportionately smaller with age. Thalamic neuron receptive fields become more multiwhisker, whereas those of cortical neurons become more single-whisker. Thalamic neurons develop a higher degree of angular selectivity, whereas that of cortical neurons remains constant. In the temporal domain, response latencies decrease both in thalamic and cortical neurons, but the maturation time-course differs between the two populations. Response latencies of thalamic cells decrease primarily between 2 and 3 wk of life, whereas response latencies of cortical neurons decrease in two distinct steps--the first between 2 and 3 wk of life and the second between the fourth postnatal week and adulthood. Although the first step likely reflects similar subcortical changes, the second phase likely corresponds to developmental myelination of thalamocortical fibers. Divergent development of thalamic and cortical response properties indicates that thalamocortical circuits in the whisker-to-barrel pathway undergo protracted maturation after 2 wk of life and provides a potential substrate for experience-dependent plasticity during this time.
Collapse
Affiliation(s)
- Michael Shoykhet
- Department of Neurobiology, Univiversity of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | | |
Collapse
|
40
|
He Y, Dupree J, Wang J, Sandoval J, Li J, Liu H, Shi Y, Nave KA, Casaccia-Bonnefil P. The transcription factor Yin Yang 1 is essential for oligodendrocyte progenitor differentiation. Neuron 2007; 55:217-30. [PMID: 17640524 PMCID: PMC2034312 DOI: 10.1016/j.neuron.2007.06.029] [Citation(s) in RCA: 213] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2006] [Revised: 05/24/2007] [Accepted: 06/21/2007] [Indexed: 11/17/2022]
Abstract
The progression of progenitors to oligodendrocytes requires proliferative arrest and the activation of a transcriptional program of differentiation. While regulation of cell cycle exit has been extensively characterized, the molecular mechanisms responsible for the initiation of differentiation remain ill-defined. Here, we identify the transcription factor Yin Yang 1 (YY1) as a critical regulator of oligodendrocyte progenitor differentiation. Conditional ablation of yy1 in the oligodendrocyte lineage in vivo induces a phenotype characterized by defective myelination, ataxia, and tremor. At the cellular level, lack of yy1 arrests differentiation of oligodendrocyte progenitors after they exit from the cell cycle. At the molecular level, YY1 acts as a lineage-specific repressor of transcriptional inhibitors of myelin gene expression (Tcf4 and Id4), by recruiting histone deacetylase-1 to their promoters during oligodendrocyte differentiation. Thus, we identify YY1 as an essential component of the transcriptional network regulating the transition of oligodendrocyte progenitors from cell cycle exit to differentiation.
Collapse
Affiliation(s)
- Ye He
- Department of Neuroscience and Cell Biology, R. Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Henkel CK, Keiger CJ, Franklin SR, Brunso-Bechtold JK. Development of banded afferent compartments in the inferior colliculus before onset of hearing in ferrets. Neuroscience 2007; 146:225-35. [PMID: 17324524 PMCID: PMC1973092 DOI: 10.1016/j.neuroscience.2007.01.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2006] [Revised: 01/05/2007] [Accepted: 01/12/2007] [Indexed: 11/28/2022]
Abstract
Axonal projections from the lateral superior olivary nuclei (LSO), as well as from the dorsal cochlear nucleus (DCN) and dorsal nucleus of the lateral lemniscus (DNLL), converge in frequency-ordered layers in the central nucleus of the inferior colliculus (IC) where they distribute among different synaptic compartments. A carbocyanine dye, 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI), was used as a tracer to study the postnatal development of axonal projections in the ferret IC. The results indicated that projections from all three nuclei are present at birth, but are not segregated into bands. During the postnatal week between approximately postnatal days 4 and 12 (P4-P12), axons from LSO proliferate in IC, become more branched, and segregate into a series of bands composed of densely packed fibers and endings. LSO projections in these afferent bands course parallel to IC layers and are separated by intervening regions with few endings. A modest fit of a sine curve (R2>0.15) to the pattern of spacing of LSO projections in IC indicated that regularly spaced bands are forming by P7. Similarly, banded patterns of DCN and DNLL projections to IC have developed by the end of the first postnatal week. Thus, well before hearing onset in ferret (P28-30), three different afferent projections have segregated into banded compartments along layers in the central nucleus of the ferret IC. Possible mechanisms in circuit development are discussed.
Collapse
Affiliation(s)
- C K Henkel
- Wake Forest University Health Sciences, Neuroscience Program and Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA.
| | | | | | | |
Collapse
|
42
|
Milh M, Becq H, Villeneuve N, Ben-Ari Y, Aniksztejn L. Inhibition of Glutamate Transporters Results in a "Suppression-Burst" Pattern and Partial Seizures in the Newborn Rat. Epilepsia 2007; 48:169-74. [PMID: 17241224 DOI: 10.1111/j.1528-1167.2006.00839.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE To determine the electrophysiological pattern and propose a clinical relevance of a deficient glutamate transport in the developing brain. METHODS (a) Surface EEG-video monitoring in freely moving pups; (b) intracortical multiple unit activity (MUA) and local field potential recordings in 5- to 7-day-old rats after pharmacological inhibition of the glutamate transporters by DL-TBOA. RESULTS Glutamate transporters inhibition alters the background cortical electrical activity inducing a dominant and persistent pattern of bilateral recurrent paroxysmal bursts alternating with periods of hypoactivity and also partial seizures. Intracortical local field recordings show that paroxysmal bursts are associated with multiunits and gamma oscillations separated by periods of silence. This cortical activity involves the activation of ionotropic glutamate receptors and was not observed after kainate and pilocarpine administration. CONCLUSIONS We show that a dysfunction of glutamate transporters in immature rats leads to a singular cortical activity that is reminiscent of a "suppression-burst" pattern. We propose that an early deficiency of glutamate transport may underlie some early onset epilepsies.
Collapse
Affiliation(s)
- Mathieu Milh
- INMED/INSERM U29, Université de la Mediterranée, Marseille, France
| | | | | | | | | |
Collapse
|
43
|
Yang P, Baker KA, Hagg T. The ADAMs family: coordinators of nervous system development, plasticity and repair. Prog Neurobiol 2006; 79:73-94. [PMID: 16824663 DOI: 10.1016/j.pneurobio.2006.05.001] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2006] [Revised: 05/18/2006] [Accepted: 05/23/2006] [Indexed: 10/24/2022]
Abstract
A disintegrin and metalloprotease (ADAM) transmembrane proteins have metalloprotease, integrin-binding, intracellular signaling and cell adhesion activities. In contrast to other metalloproteases, ADAMs are particularly important for cleavage-dependent activation of proteins such as Notch, amyloid precursor protein (APP) and transforming growth factor alpha (TGFalpha), and can bind integrins. Not surprisingly, ADAMs have been shown or suggested to play important roles in the development of the nervous system, where they regulate proliferation, migration, differentiation and survival of various cells, as well as axonal growth and myelination. On the eleventh anniversary of the naming of this family of proteins, the relatively unknown ADAMs are emerging as potential therapeutic targets for neural repair. For example, over-expression of ADAM10, one of the alpha-secretases for APP, can prevent amyloid formation and hippocampal defects in an Alzheimer mouse model. Another example of this potential neural repair role is the finding that ADAM21 is uniquely associated with neurogenesis and growing axons of the adult brain. This comprehensive review will discuss the growing literature about the roles of ADAMs in the developing and adult nervous system, and their potential roles in neurological disorders. Most excitingly, the expanding understanding of their normal roles suggests that they can be manipulated to promote neural repair in the degenerating and injured adult nervous system.
Collapse
Affiliation(s)
- Peng Yang
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville, Louisville, KY 40292, USA
| | | | | |
Collapse
|
44
|
Jeon SW, Shure MA, Baker KB, Huang D, Rollins AM, Chahlavi A, Rezai AR. A feasibility study of optical coherence tomography for guiding deep brain probes. J Neurosci Methods 2006; 154:96-101. [PMID: 16480773 PMCID: PMC1769312 DOI: 10.1016/j.jneumeth.2005.12.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2005] [Accepted: 12/05/2005] [Indexed: 11/20/2022]
Abstract
Deep brain simulation (DBS) is effective for the treatment of various diseases including Parkinson's disease and essential tremor. However, anatomical targeting combined with microelectrode mapping of the region requires significant surgical time. Also, the fine-tipped microelectrode imposes a risk of hemorrhage in the event that the trajectory intersects subcortical vessels. To reduce the operation time and the risk of hemorrhage, we propose to use optical coherence tomography (OCT) to guide the insertion of the DBS probe. We conducted in vitro experiments in the rat brain to study the feasibility of this application. The result shows that OCT is able to differentiate structures in the rat brain. White matter tends to have higher peak reflectivity and steeper attenuation rate compared to gray matter. This structural information may help guide DBS probe advance and electrical measurements.
Collapse
Affiliation(s)
- Sung W Jeon
- Center for Neurological Restoration, Cleveland Clinic Foundation, OH 44195, USA.
| | | | | | | | | | | | | |
Collapse
|
45
|
York RG, Barnett J, Girard MF, Mattie DR, Bekkedal MVK, Garman RH, Strawson JS. Refining the effects observed in a developmental neurobehavioral study of ammonium perchlorate administered orally in drinking water to rats. II. Behavioral and neurodevelopment effects. Int J Toxicol 2006; 24:451-67. [PMID: 16393938 DOI: 10.1080/10915810500367094] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
A developmental neurotoxicity study was conducted to generate additional data on the potential functional and morphological hazard to the central nervous system caused by ammonium perchlorate in offspring from in utero and lactation exposure. Female Sprague-Dawley rats (23 to 25/group) were given continuous access to 0 (carrier), 0.1, 1.0, 3.0, and 10.0 mg/kg-day perchlorate in the drinking water beginning 2 weeks prior to mating and continuing through day 10 of lactation for the behavioral function assessment or given continuous access to 0 (carrier), 0.1, 1.0, 3.0, and 30.0 mg/kg-day beginning on gestation day 0 and continuing through day 10 of lactation for neurodevelopment assessments. Motor activity was conducted on postpartum days 14, 18, and 22 and juvenile brain weights, neurohistopathological examinations, and regional brain morphometry were conducted on postpartum days 10 and 22. This research revealed a sexually dimorphic response, with some brain regions being larger in perchlorate-treated male rats than in comparable controls. Even so, there was no evidence of any obvious exposure-related effects on male rat brain weights or neuropathology. The most consistent exposure-related effect in the male pups was on the thickness of the corpus callosum, with both the right- and left-sided measures of the thickness of this white matter tract being significantly greater for the male pups in the 0.1 and 1.0 mg/kg-day exposure groups. The behavioral testing suggests prenatal exposure to ammonium perchlorate does not affect the development of gross motor movements in the pups.
Collapse
Affiliation(s)
- Raymond G York
- Charles River Laboratories, Preclinical Services, 905 Sheehy Drive, Horsham, PA 19044, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
Kaur C, Sivakumar V, Dheen ST, Ling EA. Insulin-like growth factor I and II expression and modulation in amoeboid microglial cells by lipopolysaccharide and retinoic acid. Neuroscience 2006; 138:1233-44. [PMID: 16448778 DOI: 10.1016/j.neuroscience.2005.12.025] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2005] [Revised: 11/23/2005] [Accepted: 12/01/2005] [Indexed: 01/15/2023]
Abstract
Insulin-like growth factors I and II are known to regulate the development of the CNS. We examined the developmental changes in insulin-like growth factor I and insulin-like growth factor II expression in the postnatal rat corpus callosum. Insulin-like growth factor I and insulin-like growth factor II mRNA expression increased at 3 days as compared with 1 day whereas the protein expression increased up to 7 days. Insulin-like growth factor I and insulin-like growth factor II immunoexpression was specifically localized in round cells confirmed by double immunofluorescence with OX-42 to be the amoeboid microglial cells. Insulin-like growth factor I expression was observed up to 7 days in amoeboid microglial cells while insulin-like growth factor II expression was detected in 1-3 day old rats. Exposure of primary rat microglial cell cultures to lipopolysaccharide increased insulin-like growth factor I and insulin-like growth factor II mRNA and protein expression significantly along with their immunoexpression in microglial cells. The lipopolysaccharide-induced increase in insulin-like growth factor I and insulin-like growth factor II mRNA and protein expression was significantly decreased with all-trans-retinoic acid. We conclude that insulin-like growth factor I and insulin-like growth factor II expression in amoeboid microglial cells in the developing brain is related to their activation. Once the activation is inhibited, either by transformation of the amoeboid microglial cells into ramified microglia regarded as resting cells or as shown by the effect of all-trans-retinoic acid administration, insulin-like growth factor I and insulin-like growth factor II mRNA and protein expression is downregulated.
Collapse
Affiliation(s)
- C Kaur
- Department of Anatomy, Yong Loo Lin School of Medicine, Blk MD10, 4 Medical Drive, National University of Singapore, Singapore 117597.
| | | | | | | |
Collapse
|
47
|
Zhang B, Cao Q, Guo A, Chu H, Chan YG, Buschdorf JP, Low BC, Ling EA, Liang F. Juxtanodin: an oligodendroglial protein that promotes cellular arborization and 2',3'-cyclic nucleotide-3'-phosphodiesterase trafficking. Proc Natl Acad Sci U S A 2005; 102:11527-32. [PMID: 16051705 PMCID: PMC1183540 DOI: 10.1073/pnas.0500952102] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
In the process of screening cell-type-specific genes, we identified juxtanodin (JN), an oligodendroglial protein featuring a putative C-terminal actin-binding domain. At the cellular level, JN in the rat CNS colocalized with 2',3'-cyclic nucleotide-3'-phosphodiesterase (CNPase), a cytoskeleton-related oligodendroglial protein. In the myelin sheath, JN was found mainly in the abaxon and the lateral few terminal loops. Its apposition to the myelinated axon, through the latter, defined an axonal subregion, herewith termed juxtanode, at the Ranvier node-paranode junction. During forebrain ontogenesis, JN expression paralleled that of MBPs but lagged behind CNPase. Juxtanodin transfection promoted arborization of cultured OLN-93 cells and augmented endogenous CNPase expression and transport to the process arbors of cultured primary oligodendrocyte precursors. These results reveal JN as a cytoskeleton-related oligodendroglial protein that delineates the juxtanode and might serve oligodendrocyte motility, differentiation, or myelin-axon signaling. Functionally, JN may be involved in CNS myelination and/or specialization of the node of Ranvier.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Anatomy, Faculty of Medicine, National University of Singapore, Singapore 117597
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Shen S, Li J, Casaccia-Bonnefil P. Histone modifications affect timing of oligodendrocyte progenitor differentiation in the developing rat brain. ACTA ACUST UNITED AC 2005; 169:577-89. [PMID: 15897262 PMCID: PMC2171688 DOI: 10.1083/jcb.200412101] [Citation(s) in RCA: 247] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Timely differentiation of progenitor cells is critical for development. In this study we asked whether global epigenetic mechanisms regulate timing of progenitor cell differentiation into myelin-forming oligodendrocytes in vivo. Histone deacetylation was essential during a specific temporal window of development and was dependent on the enzymatic activity of histone deacetylases, whose expression was detected in the developing corpus callosum. During the first 10 postnatal days, administration of valproic acid (VPA), the specific inhibitor for histone deacetylase activity, resulted in significant hypomyelination with delayed expression of late differentiation markers and retained expression of progenitor markers. Differentiation resumed in VPA-injected rats if a recovery period was allowed. Administration of VPA after myelination onset had no effect on myelin gene expression and was consistent with changes of nucleosomal histones from reversible deacetylation to more stable methylation and chromatin compaction. Together, these data identify global modifications of nucleosomal histones critical for timing of oligodendrocyte differentiation and myelination in the developing corpus callosum.
Collapse
Affiliation(s)
- Siming Shen
- Department of Neuroscience and Cell Biology, R. Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | | | |
Collapse
|
49
|
Bernstein HG, Keilhoff G, Bukowska A, Ziegeler A, Funke S, Dobrowolny H, Kanakis D, Bogerts B, Lendeckel U. ADAM (a disintegrin and metalloprotease) 12 is expressed in rat and human brain and localized to oligodendrocytes. J Neurosci Res 2004; 75:353-60. [PMID: 14743448 DOI: 10.1002/jnr.10858] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
ADAM12 is a member of the large family of multidomain metalloprotease-disintegrins which possess cell-binding and metalloprotease properties. Typically, ADAM12 is expressed in mesenchymal cells, developing and regenerating heart and skeletal muscle, bone as well as in certain tumours. This report shows by means of reverse transcriptase-polymerase chain reaction (RT-PCR) and immunohistochemistry that the protease ADAM12 is detectable in human and rat brain tissue as well as in cultured cells derived from rat brain. With the exception of a very few immunopositive pyramidal neurons in the developing rat brain, the cellular localization of ADAM12 was exclusively confined to oligodendroglial cells. Thus, ADAM12 may be regarded a new suitable marker for this cell type.
Collapse
|
50
|
Nuñez JL, Nelson J, Pych JC, Kim JH, Juraska JM. Myelination in the splenium of the corpus callosum in adult male and female rats. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2000; 120:87-90. [PMID: 10727734 DOI: 10.1016/s0165-3806(99)00193-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Previous work reported increases in the number of myelinated axons in the splenium of the rat corpus callosum between 25 and 60 days of age. In the present study, we quantified the area occupied by myelinated axons using a light microscopic point counting technique at 60, 120 and 180 days. Myelinated axons increased across these ages (p=0.001). Thus, myelination of the rat corpus callosum persists well into adulthood.
Collapse
Affiliation(s)
- J L Nuñez
- Neuroscience Program and Department of Psychology, University of Illinois at Champaign-Urbana, 603 E Daniel Street, Champaign, IL, USA
| | | | | | | | | |
Collapse
|