1
|
Tang Y, Gutmann DH. Neurofibromatosis Type 1-Associated Optic Pathway Gliomas: Current Challenges and Future Prospects. Cancer Manag Res 2023; 15:667-681. [PMID: 37465080 PMCID: PMC10351533 DOI: 10.2147/cmar.s362678] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/06/2023] [Indexed: 07/20/2023] Open
Abstract
Optic pathway glioma (OPG) occurs in as many as one-fifth of individuals with the neurofibromatosis type 1 (NF1) cancer predisposition syndrome. Generally considered low-grade and slow growing, many children with NF1-OPGs remain asymptomatic. However, due to their location within the optic pathway, ~20-30% of those harboring NF1-OPGs will experience symptoms, including progressive vision loss, proptosis, diplopia, and precocious puberty. While treatment with conventional chemotherapy is largely effective at attenuating tumor growth, it is not clear whether there is much long-term recovery of visual function. Additionally, because these tumors predominantly affect young children, there are unique challenges to NF1-OPG diagnosis, monitoring, and longitudinal management. Over the past two decades, the employment of authenticated genetically engineered Nf1-OPG mouse models have provided key insights into the function of the NF1 protein, neurofibromin, as well as the molecular and cellular pathways that contribute to optic gliomagenesis. Findings from these studies have resulted in the identification of new molecular targets whose inhibition blocks murine Nf1-OPG growth in preclinical studies. Some of these promising compounds have now entered into early clinical trials. Future research focused on defining the determinants that underlie optic glioma initiation, expansion, and tumor-induced optic nerve injury will pave the way to personalized risk assessment strategies, improved tumor monitoring, and optimized treatment plans for children with NF1-OPG.
Collapse
Affiliation(s)
- Yunshuo Tang
- Department of Ophthalmology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
2
|
Ono K, Gotoh H, Nomura T, Morita T, Baba O, Matsumoto M, Saitoh S, Ohno N. Ultrastructural characteristics of oligodendrocyte precursor cells in the early postnatal mouse optic nerve observed by serial block-face scanning electron microscopy. PLoS One 2022; 17:e0278118. [PMID: 36454994 PMCID: PMC9714907 DOI: 10.1371/journal.pone.0278118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/10/2022] [Indexed: 12/05/2022] Open
Abstract
Oligodendrocyte precursor cells (OPC) arise from restricted regions of the central nervous system (CNS) and differentiate into myelin-forming cells after migration, but their ultrastructural characteristics have not been fully elucidated. This study examined the three-dimensional ultrastructure of OPCs in comparison with other glial cells in the early postnatal optic nerve by serial block-face scanning electron microscopy. We examined 70 putative OPCs (pOPC) that were distinct from other glial cells according to established morphological criteria. The pOPCs were unipolar in shape with relatively few processes, and their Golgi apparatus were localized in the perinuclear region with a single cisterna. Astrocytes abundant in the optic nerve were distinct from pOPCs and had a greater number of processes and more complicated Golgi apparatus morphology. All pOPCs and astrocytes contained a pair of centrioles (basal bodies). Among them, 45% of pOPCs extended a short cilium, and 20% of pOPCs had centrioles accompanied by vesicles, whereas all astrocytes with basal bodies had cilia with invaginated ciliary pockets. These results suggest that the fine structures of pOPCs during the developing and immature stages may account for their distinct behavior. Additionally, the vesicular transport of the centrioles, along with a short cilium length, suggests active ciliogenesis in pOPCs.
Collapse
Affiliation(s)
- Katsuhiko Ono
- Developmental Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hitoshi Gotoh
- Developmental Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tadashi Nomura
- Developmental Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tsuyoshi Morita
- Oral & Maxillofacial Anatomy, Graduate School of Oral Science, Tokushima University, Tokushima, Japan
| | - Otto Baba
- Oral & Maxillofacial Anatomy, Graduate School of Oral Science, Tokushima University, Tokushima, Japan
| | - Mami Matsumoto
- Section of Electron Microscopy, Supportive for Brain Research, National Institute for Physiological Sciences, Okazaki, Japan
- Developmental & Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Sei Saitoh
- Section of Electron Microscopy, Supportive for Brain Research, National Institute for Physiological Sciences, Okazaki, Japan
- Department of Anatomy II and Cell Biology, Fujita Health University, School of Medicine, Toyoake, Japan
| | - Nobuhiko Ohno
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine, Tochigi, Japan
- Division of Ultrastructural Research, National Institute for Physiological Sciences, Okazaki, Japan
| |
Collapse
|
3
|
Endogenous Neural Stem Cell Mediated Oligodendrogenesis in the Adult Mammalian Brain. Cells 2022; 11:cells11132101. [PMID: 35805185 PMCID: PMC9265817 DOI: 10.3390/cells11132101] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 02/08/2023] Open
Abstract
Oligodendrogenesis is essential for replacing worn-out oligodendrocytes, promoting myelin plasticity, and for myelin repair following a demyelinating injury in the adult mammalian brain. Neural stem cells are an important source of oligodendrocytes in the adult brain; however, there are considerable differences in oligodendrogenesis from neural stem cells residing in different areas of the adult brain. Amongst the distinct niches containing neural stem cells, the subventricular zone lining the lateral ventricles and the subgranular zone in the dentate gyrus of the hippocampus are considered the principle areas of adult neurogenesis. In addition to these areas, radial glia-like cells, which are the precursors of neural stem cells, are found in the lining of the third ventricle, where they are called tanycytes, and in the cerebellum, where they are called Bergmann glia. In this review, we will describe the contribution and regulation of each of these niches in adult oligodendrogenesis.
Collapse
|
4
|
D'Angelo F, Lasorella A. Inhibition of ERK/MAPK signaling as potential therapy to prevent optic pathway glioma in infants with neurofibromatosis type 1. Dev Cell 2021; 56:2785-2786. [PMID: 34699786 DOI: 10.1016/j.devcel.2021.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Pediatric low-grade gliomas (pLGGs) arise primarily at early stages of development. The molecular mechanisms of pLGG gliomagenesis are unclear, as is the progenitor cell of origin. In this issue of Developmental Cell, Jecrois et al. show that NF1-associated optic pathway gliomas originate from migrating glial progenitors that have distinct MEK/ERK dependency.
Collapse
Affiliation(s)
- Fulvio D'Angelo
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY 10032, USA
| | - Anna Lasorella
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA; Department of Pediatrics, Columbia University Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
5
|
Yamagata M, Yan W, Sanes JR. A cell atlas of the chick retina based on single-cell transcriptomics. eLife 2021; 10:e63907. [PMID: 33393903 PMCID: PMC7837701 DOI: 10.7554/elife.63907] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/01/2021] [Indexed: 12/14/2022] Open
Abstract
Retinal structure and function have been studied in many vertebrate orders, but molecular characterization has been largely confined to mammals. We used single-cell RNA sequencing (scRNA-seq) to generate a cell atlas of the chick retina. We identified 136 cell types plus 14 positional or developmental intermediates distributed among the six classes conserved across vertebrates - photoreceptor, horizontal, bipolar, amacrine, retinal ganglion, and glial cells. To assess morphology of molecularly defined types, we adapted a method for CRISPR-based integration of reporters into selectively expressed genes. For Müller glia, we found that transcriptionally distinct cells were regionally localized along the anterior-posterior, dorsal-ventral, and central-peripheral retinal axes. We also identified immature photoreceptor, horizontal cell, and oligodendrocyte types that persist into late embryonic stages. Finally, we analyzed relationships among chick, mouse, and primate retinal cell classes and types. Our results provide a foundation for anatomical, physiological, evolutionary, and developmental studies of the avian visual system.
Collapse
Affiliation(s)
- Masahito Yamagata
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard UniversityCambridgeUnited States
| | - Wenjun Yan
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard UniversityCambridgeUnited States
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard UniversityCambridgeUnited States
| |
Collapse
|
6
|
Boonzaier NR, Hales PW, D'Arco F, Walters BC, Kaur R, Mankad K, Cooper J, Liasis A, Smith V, O'Hare P, Hargrave D, Clark CA. Quantitative MRI demonstrates abnormalities of the third ventricle subventricular zone in neurofibromatosis type-1 and sporadic paediatric optic pathway glioma. NEUROIMAGE-CLINICAL 2020; 28:102447. [PMID: 33038669 PMCID: PMC7554210 DOI: 10.1016/j.nicl.2020.102447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 09/17/2020] [Accepted: 09/20/2020] [Indexed: 11/26/2022]
Abstract
MRI provides supporting evidence of third ventricle subventricular involvement in OPG. Third ventricle subventricular zone ADC and CBF differs between NF1 and sporadic OPG. Third ventricle subventricular zone ADC correlates with vision in sporadic OPG.
Background The subventricular zone of the third ventricle (TVZ) is a germinal stem cell niche, identified as the possible location of optic pathway glioma (OPG) cell origin. Paediatric OPGs are predominantly diagnosed as low-grade astrocytomas, which are either sporadic or are associated with neurofibromatosis type-1 (NF1). These tumours often cause a significant impairment to visual acuity (VA). Infiltrative/invasive tumour activity is associated with increased apparent diffusion coefficient (ADC) and cerebral blood flow (CBF). This study aimed to determine whether TVZ imaging features differed between sporadic-OPG, NF1-OPG and controls, and whether the ADC and CBF profile at the germinal stem cell niche (the TVZ) correlated with the primary outcome of VA. Methods ADC and CBF MRI data were acquired from 30 paediatric OPG patients (median age 6 years; range 8 months–17 years), along with VA measurements, during clinical surveillance of their tumour. Values for mean ADC and maximum CBF were measured at the TVZ, and normalized to normal-appearing grey matter. These values were compared between the two OPG groups and the healthy control subjects, and multivariate linear regression was used to test the linear association between these values and patient’s VA. Results In the TVZ, normalized mean ADC was higher in NF1-associated OPG patients (N = 15), compared to both sporadic OPG patients (N = 15; p = 0.010) and healthy controls (N = 14; p < 0.001). In the same region, normalized maximum CBF was higher in sporadic OPG patients compared to both NF1-OPG patients (p = 0.016) and healthy controls (p < 0.001). In sporadic OPG patients only, normalized mean ADC in the TVZ was significantly correlated with visual acuity (R2 = 0.41, p = 0.019). No significant correlations were found between TVZ CBF and ADC values and visual acuity in the NF1-associated OPG patients. Conclusion Quantitative MRI detects TVZ abnormalities in both sporadic and NF1-OPG patients, and identifies TVZ features that differentiate the two. TVZ features may be useful MRI markers of interest in future predictive studies involving sporadic OPG.
Collapse
Affiliation(s)
- Natalie R Boonzaier
- Developmental Imaging and Biophysics Section, Developmental Neurosciences, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Patrick W Hales
- Developmental Imaging and Biophysics Section, Developmental Neurosciences, University College London Great Ormond Street Institute of Child Health, London, UK.
| | - Felice D'Arco
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Bronwen C Walters
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Ramneek Kaur
- Developmental Imaging and Biophysics Section, Developmental Neurosciences, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Kshitij Mankad
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Jessica Cooper
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Alki Liasis
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK; University of Pittsburgh Medical Center, Children's Hospital of Pittsburgh, Pittsburgh, USA
| | - Victoria Smith
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Patricia O'Hare
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Darren Hargrave
- Developmental Imaging and Biophysics Section, Developmental Neurosciences, University College London Great Ormond Street Institute of Child Health, London, UK; Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Christopher A Clark
- Developmental Imaging and Biophysics Section, Developmental Neurosciences, University College London Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
7
|
Latini F, Fahlström M, Hesselager G, Zetterling M, Ryttlefors M. Differences in the preferential location and invasiveness of diffuse low-grade gliomas and their impact on outcome. Cancer Med 2020; 9:5446-5458. [PMID: 32537906 PMCID: PMC7402839 DOI: 10.1002/cam4.3216] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/14/2020] [Accepted: 05/16/2020] [Indexed: 12/16/2022] Open
Abstract
Background Low‐grade gliomas (LGGs) are primary diffuse slow‐growing brain tumors derived from glial cells. The management of these tumors is dependent on their location, which often harbors eloquent areas. We retrospectively recorded the location of diffuse gliomas to identify whether specific differences exist between the histological types. Methods We analyzed 102 patients with previous histological diagnosis of WHO‐II astrocytomas (62) and WHO‐II oligodendrogliomas (40) according to WHO‐2016 classification. MRI sequences (T2‐FLAIR) were used for tumor volume segmentation and to create a frequency map of their locations within the Montreal Neurological Institute (MNI) space. The Brain‐Grid (BG) system (standardized radiological tool of intersected lines according to anatomical landmarks) was created and merged with a tractography atlas for infiltration analysis. Results Astrocytomas frequently infiltrated association and projection white matter pathways within fronto‐temporo‐insular regions on the left side. Oligodendrogliomas infiltrated larger white matter networks (association‐commissural‐projection) of the frontal lobe bilaterally. A critical number of infiltrated BG voxels (7 for astrocytomas, 10 for oligodendrogliomas) significantly predicted shorter overall survival (OS) in both groups. Bilateral tumor extension in astrocytomas and preoperative tumor volume in oligodendrogliomas were independent prognostic factors for shorter OS. Conclusions Astrocytomas and oligodendrogliomas differ in preferential location, and this has an impact on the type and the extent of white matter involvement. The number of BG voxels infiltrated reflected different tumor invasiveness and its impact on OS in both groups. All this new information may be valuable in neurosurgical oncology to classify and plan treatment for patients with diffuse gliomas.
Collapse
Affiliation(s)
- Francesco Latini
- Department of Neuroscience, Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Markus Fahlström
- Department of Surgical Sciences, Radiology, Uppsala University, Uppsala, Sweden
| | - Göran Hesselager
- Department of Neuroscience, Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Maria Zetterling
- Department of Neuroscience, Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Mats Ryttlefors
- Department of Neuroscience, Neurosurgery, Uppsala University, Uppsala, Sweden
| |
Collapse
|
8
|
Swiatczak B, Feldkaemper M, Schraermeyer U, Schaeffel F. Demyelination and shrinkage of axons in the retinal nerve fiber layer in chickens developing deprivation myopia. Exp Eye Res 2019; 188:107783. [PMID: 31473258 DOI: 10.1016/j.exer.2019.107783] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/16/2019] [Accepted: 08/29/2019] [Indexed: 12/30/2022]
Abstract
Placing diffusers in front of the eyes induces deprivation myopia in a variety of animal models. As a result of the low pass filtering of the retinal images, less spatial information is available to the retina which should reduce neural activity. Since it has been found that myelination of axons in the central nervous system is modulated by neuronal activity, we have studied whether ganglion cell axons may shrink in response to the restricted visual input. Young chickens were treated for 5 h or 7 days with frosted diffusers to induce deprivation myopia. Nerve fiber layer thickness was measured in vivo, using B-scan OCT. Refractive states were tracked by IR photoretinoscopy, and UV fundus reflectivity by a custom-built device which flashed an LED centered in the camera aperture and recorded pupil brightness after refractive errors were corrected by trial lenses. Moreover, structure and histology of the retinal nerve fibers layer (RNFL) were analyzed ex vivo using transmission electron microscopy and immunohistochemistry. Since chicks have both non-myelinated and myelinated fibers in their RNFL, the thickness of myelin sheaths (G ratio) was measured, as well as the percentage of myelinated axons and the diameters of unmyelinated axons. Short-term deprivation caused an increase in UV fundus reflectivity already after 5 h (measured as pixel grey levels in the pupil: 28 ± 5 vs. 36 ± 10, p < 0.05) and thinning of the myelin sheaths (higher G ratio), compared to untreated control eyes (0.74 ± 0.01 vs. 0.79 ± 0.03, p < 0.05). Neither axon diameters (0.81 ± 0.05 μm vs. 0.82 ± 0.15 μm) nor thickness of the RNFL had changed after only 5 h (42.9 ± 1.3 μm vs. 42.3 ± 2.5 μm). However, after 7 days of diffuser wear, axons had become thinner (0.56 ± 0.14 μm vs. 0.78 ± 0.09 μm vs, p < 0.05), which could explain the thinning of the RNFL (36.3 ± 2.7 μm vs. 42.1 ± 2.4 μm, p < 0.01). Furthermore, myopic eyes had 38% less myelinated axons than untreated eyes as determined by immunohistochemical labelling against myelin basic protein (immunopositive areas in the central retina 1406 ± 341 μm2 vs. 2185 ± 290 μm2 in controls, p < 0.001). Myelin sheaths in the remaining axons remained unchanged (G ratio 0.76 ± 0.02 vs. 0.76 ± 0.03). Our study shows that deprivation myopia is associated with a significant loss of myelinated axons and shrinkage of the axon diameters of certain fibers in the RNFL. Early changes were already detected after 5 h and were accompanied by an increased fundus reflectivity in UV light. These parameters could therefore serve as the biomarkers for myopia development, at least in the chicken.
Collapse
Affiliation(s)
- Barbara Swiatczak
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, Elfriede Aulhorn Str. 7, 72076, Tuebingen, Germany
| | - Marita Feldkaemper
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, Elfriede Aulhorn Str. 7, 72076, Tuebingen, Germany
| | - Ulrich Schraermeyer
- Experimental Vitreoretinal Surgery, Ophthalmic Research Institute, University of Tuebingen, Schleichstr. 12/1, 72076, Tuebingen, Germany
| | - Frank Schaeffel
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, Elfriede Aulhorn Str. 7, 72076, Tuebingen, Germany.
| |
Collapse
|
9
|
Pajoohesh-Ganji A, Miller RH. Targeted Oligodendrocyte Apoptosis in Optic Nerve Leads to Persistent Demyelination. Neurochem Res 2019; 45:580-590. [PMID: 30848441 PMCID: PMC7058578 DOI: 10.1007/s11064-019-02754-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/11/2019] [Accepted: 02/12/2019] [Indexed: 01/06/2023]
Abstract
The optic nerve represents one of the simplest regions of the CNS and has been useful in developing an understanding of glial development and myelination. While the visual system is frequently affected in demyelinating conditions, utilizing the optic nerve to model demyelination/remyelination studies has been difficult due to its accessibility, relatively small size, and dense nature that makes direct injections challenging. Taking advantage of the lack of oligodendrocytes and myelination in the mouse retina, we have developed a model in which the induction of apoptosis in mature oligodendrocytes allows for the selective, non-invasive generation of demyelinating lesions in optic nerve. Delivery of an inducer of oligodendrocyte apoptosis by intravitreous injection minimizes trauma to the optic nerve and allows for the assessment of oligodendrocyte death in the absence of injury related factors. Here we show that following induction of apoptosis, oligodendrocytes are lost within 3 days. The loss of oligodendrocytes is associated with limited microglial and astrocyte response, is patchy along the nerve, and results in localized myelin loss. Unlike in other regions of the murine CNS, where local demyelination stimulates activation of local oligodendrocyte precursors and remyelination, optic nerve demyelination induced by oligodendrocyte apoptosis fails to recover and results in persistent areas of myelin loss. Over time these chronic lesions change cellular composition and ultimately become devoid of GFAP+ astrocytes and OPCs. Why the optic nerve lesions fail to repair may reflect the lack of early immune responsiveness and provide a novel model of chronic demyelination.
Collapse
Affiliation(s)
- Ahdeah Pajoohesh-Ganji
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA.
| | - Robert H Miller
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| |
Collapse
|
10
|
Freret ME, Gutmann DH. Insights into optic pathway glioma vision loss from mouse models of neurofibromatosis type 1. J Neurosci Res 2018; 97:45-56. [PMID: 29704429 DOI: 10.1002/jnr.24250] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 04/09/2018] [Indexed: 12/12/2022]
Abstract
Neurofibromatosis type 1 (NF1) is a common cancer predisposition syndrome caused by mutations in the NF1 gene. The NF1-encoded protein (neurofibromin) is an inhibitor of the oncoprotein RAS and controls cell growth and survival. Individuals with NF1 are prone to developing low-grade tumors of the optic nerves, chiasm, tracts, and radiations, termed optic pathway gliomas (OPGs), which can cause vision loss. A paucity of surgical tumor specimens and of patient-derived xenografts for investigative studies has limited our understanding of human NF1-associated OPG (NF1-OPG). However, mice genetically engineered to harbor Nf1 gene mutations develop optic gliomas that share many features of their human counterparts. These genetically engineered mouse (GEM) strains have provided important insights into the cellular and molecular determinants that underlie mouse Nf1 optic glioma development, maintenance, and associated vision loss, with relevance by extension to human NF1-OPG disease. Herein, we review our current understanding of NF1-OPG pathobiology and describe the mechanisms responsible for tumor initiation, growth, and associated vision loss in Nf1 GEM models. We also discuss how Nf1 GEM and other preclinical models can be deployed to identify and evaluate molecularly targeted therapies for OPG, particularly as they pertain to future strategies aimed at preventing or improving tumor-associated vision loss in children with NF1.
Collapse
Affiliation(s)
- Morgan E Freret
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
11
|
Sawada M, Ohno N, Kawaguchi M, Huang SH, Hikita T, Sakurai Y, Bang Nguyen H, Quynh Thai T, Ishido Y, Yoshida Y, Nakagawa H, Uemura A, Sawamoto K. PlexinD1 signaling controls morphological changes and migration termination in newborn neurons. EMBO J 2018; 37:embj.201797404. [PMID: 29348324 DOI: 10.15252/embj.201797404] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 10/28/2017] [Accepted: 12/15/2017] [Indexed: 12/22/2022] Open
Abstract
Newborn neurons maintain a very simple, bipolar shape, while they migrate from their birthplace toward their destinations in the brain, where they differentiate into mature neurons with complex dendritic morphologies. Here, we report a mechanism by which the termination of neuronal migration is maintained in the postnatal olfactory bulb (OB). During neuronal deceleration in the OB, newborn neurons transiently extend a protrusion from the proximal part of their leading process in the resting phase, which we refer to as a filopodium-like lateral protrusion (FLP). The FLP formation is induced by PlexinD1 downregulation and local Rac1 activation, which coincide with microtubule reorganization and the pausing of somal translocation. The somal translocation of resting neurons is suppressed by microtubule polymerization within the FLP The timing of neuronal migration termination, controlled by Sema3E-PlexinD1-Rac1 signaling, influences the final positioning, dendritic patterns, and functions of the neurons in the OB These results suggest that PlexinD1 signaling controls FLP formation and the termination of neuronal migration through a precise control of microtubule dynamics.
Collapse
Affiliation(s)
- Masato Sawada
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Nobuhiko Ohno
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Anatomy, Division of Histology and Cell Biology, Jichi Medical University, School of Medicine, Shimotsuke, Japan
| | - Mitsuyasu Kawaguchi
- Department of Organic and Medicinal Chemistry, Nagoya City University Graduate School of Pharmaceutical Sciences, Nagoya, Japan
| | - Shih-Hui Huang
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takao Hikita
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Youmei Sakurai
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Huy Bang Nguyen
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan
| | - Truc Quynh Thai
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan
| | - Yuri Ishido
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yutaka Yoshida
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Hidehiko Nakagawa
- Department of Organic and Medicinal Chemistry, Nagoya City University Graduate School of Pharmaceutical Sciences, Nagoya, Japan
| | - Akiyoshi Uemura
- Department of Retinal Vascular Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan .,Division of Neural Development and Regeneration, National Institute for Physiological Sciences, Okazaki, Japan
| |
Collapse
|
12
|
Ono K, Hirahara Y, Gotoh H, Nomura T, Takebayashi H, Yamada H, Ikenaka K. Origin of Oligodendrocytes in the Vertebrate Optic Nerve: A Review. Neurochem Res 2017; 43:3-11. [PMID: 28980095 DOI: 10.1007/s11064-017-2404-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/12/2017] [Accepted: 09/19/2017] [Indexed: 01/25/2023]
Abstract
One of the unsolved problems in the research field of oligodendrocyte (OL) development has been the site(s) of origin of optic nerve OLs and its precursor cells (OPCs). It is generally accepted that OLs in the optic nerve are derived from the brain, and thus optic nerve OLs are immigrant cells. We previously demonstrated the brain origin of optic nerve OPCs in chick embryos. However, the site of optic nerve OPC origin has not been examined experimentally in developing rodents for the past two decades. We have recently reported that optic nerve OPCs in mice arise in the preoptic area by E12.5 and gradually migrate caudally and enter the optic nerve. These OPCs give rise to myelinating OLs in the optic nerve in the postnatal or adult stages. Surprisingly, there are species differences with respect to the origin of optic nerve OPCs between chicks and mice. Here, we summarize the site of OPC origin in the optic nerve based on our own previous and recent results, and discuss possible mechanisms underlying these species differences.
Collapse
Affiliation(s)
- Katsuhiko Ono
- Developmental Neurobiology, Kyoto Prefectural University of Medicine, Kyoto, 606-0823, Japan.
| | - Yukie Hirahara
- Department of Anatomy and Cell Science, Kansai Medical University, Osaka, Japan
| | - Hitoshi Gotoh
- Developmental Neurobiology, Kyoto Prefectural University of Medicine, Kyoto, 606-0823, Japan
| | - Tadashi Nomura
- Developmental Neurobiology, Kyoto Prefectural University of Medicine, Kyoto, 606-0823, Japan
| | | | - Hisao Yamada
- Department of Anatomy and Cell Science, Kansai Medical University, Osaka, Japan
| | - Kazuhiro Ikenaka
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences (NIPS), Okazaki, Japan.,Department of Physiological Sciences, School of Life Science, The Graduate University of Advanced Studies (Sokendai), Miki-cho, Kanagawa, Japan
| |
Collapse
|
13
|
Ono K, Yoshii K, Tominaga H, Gotoh H, Nomura T, Takebayashi H, Ikenaka K. Oligodendrocyte precursor cells in the mouse optic nerve originate in the preoptic area. Brain Struct Funct 2017; 222:2441-2448. [PMID: 28293728 DOI: 10.1007/s00429-017-1394-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 02/24/2017] [Indexed: 11/28/2022]
Abstract
The present study aims to examine the origin of oligodendrocyte progenitor cells (OPCs) in the mouse optic nerve (ON) by labeling OPCs in the fetal forebrain. The labeling of OPCs in the ON was performed by injection of a retrovirus vector carrying the lacZ gene into the lateral ventricle, or by inducible Cre/loxP of Olig2-positive cells. The retrovirus labeling revealed that ventricular zone-derived cells of the fetal forebrain relocated to the ON and differentiated into oligodendrocytes. In addition, lineage tracing of Olig2-positive cells and whole-mount staining of PDGFRα-positive cells demonstrated that OPCs appeared by E12.5 in the preoptic area, and spread caudally to enter the ON. Our results also suggest that OPCs generated during the early stage are depleted from the ON after maturation.
Collapse
Affiliation(s)
- Katsuhiko Ono
- Department of Biology, Kyoto Prefectural University of Medicine, Kyoto, 606-0823, Japan.
| | - Kengo Yoshii
- Departments of Mathematics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroyuki Tominaga
- Department of Biology, Kyoto Prefectural University of Medicine, Kyoto, 606-0823, Japan
| | - Hitoshi Gotoh
- Department of Biology, Kyoto Prefectural University of Medicine, Kyoto, 606-0823, Japan
| | - Tadashi Nomura
- Department of Biology, Kyoto Prefectural University of Medicine, Kyoto, 606-0823, Japan
| | | | - Kazuhiro Ikenaka
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan
| |
Collapse
|
14
|
Dulamea AO. Role of Oligodendrocyte Dysfunction in Demyelination, Remyelination and Neurodegeneration in Multiple Sclerosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 958:91-127. [PMID: 28093710 DOI: 10.1007/978-3-319-47861-6_7] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Oligodendrocytes (OLs) are the myelinating cells of the central nervous system (CNS) during development and throughout adulthood. They result from a complex and well controlled process of activation, proliferation, migration and differentiation of oligodendrocyte progenitor cells (OPCs) from the germinative niches of the CNS. In multiple sclerosis (MS), the complex pathological process produces dysfunction and apoptosis of OLs leading to demyelination and neurodegeneration. This review attempts to describe the patterns of demyelination in MS, the steps involved in oligodendrogenesis and myelination in healthy CNS, the different pathways leading to OLs and myelin loss in MS, as well as principles involved in restoration of myelin sheaths. Environmental factors and their impact on OLs and pathological mechanisms of MS are also discussed. Finally, we will present evidence about the potential therapeutic targets in re-myelination processes that can be accessed in order to develop regenerative therapies for MS.
Collapse
Affiliation(s)
- Adriana Octaviana Dulamea
- Neurology Clinic, University of Medicine and Pharmacy "Carol Davila", Fundeni Clinical Institute, Building A, Neurology Clinic, Room 201, 022328, Bucharest, Romania.
| |
Collapse
|
15
|
Parrilla M, León-Lobera F, Lillo C, Arévalo R, Aijón J, Lara JM, Velasco A. Sox10 Expression in Goldfish Retina and Optic Nerve Head in Controls and after the Application of Two Different Lesion Paradigms. PLoS One 2016; 11:e0154703. [PMID: 27149509 PMCID: PMC4858161 DOI: 10.1371/journal.pone.0154703] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/18/2016] [Indexed: 12/24/2022] Open
Abstract
The mammalian central nervous system (CNS) is unable to regenerate. In contrast, the CNS of fish, including the visual system, is able to regenerate after damage. Moreover, the fish visual system grows continuously throughout the life of the animal, and it is therefore an excellent model to analyze processes of myelination and re-myelination after an injury. Here we analyze Sox10+ oligodendrocytes in the goldfish retina and optic nerve in controls and after two kinds of injuries: cryolesion of the peripheral growing zone and crushing of the optic nerve. We also analyze changes in a major component of myelin, myelin basic protein (MBP), as a marker for myelinated axons. Our results show that Sox10+ oligodendrocytes are located in the retinal nerve fiber layer and along the whole length of the optic nerve. MBP was found to occupy a similar location, although its loose appearance in the retina differed from the highly organized MBP+ axon bundles in the optic nerve. After optic nerve crushing, the number of Sox10+ cells decreased in the crushed area and in the optic nerve head. Consistent with this, myelination was highly reduced in both areas. In contrast, after cryolesion we did not find changes in the Sox10+ population, although we did detect some MBP- degenerating areas. We show that these modifications in Sox10+ oligodendrocytes are consistent with their role in oligodendrocyte identity, maintenance and survival, and we propose the optic nerve head as an excellent area for research aimed at better understanding of de- and remyelination processes.
Collapse
Affiliation(s)
- Marta Parrilla
- Instituto de Neurociencias de Castilla y León, Universidad de Salamanca, Salamanca, Spain
| | - Fernando León-Lobera
- Instituto de Neurociencias de Castilla y León, Universidad de Salamanca, Salamanca, Spain
- IBSAL, Salamanca, Spain
| | - Concepción Lillo
- Instituto de Neurociencias de Castilla y León, Universidad de Salamanca, Salamanca, Spain
- IBSAL, Salamanca, Spain
| | - Rosario Arévalo
- Instituto de Neurociencias de Castilla y León, Universidad de Salamanca, Salamanca, Spain
- IBSAL, Salamanca, Spain
| | - José Aijón
- Instituto de Neurociencias de Castilla y León, Universidad de Salamanca, Salamanca, Spain
- IBSAL, Salamanca, Spain
| | - Juan Manuel Lara
- Instituto de Neurociencias de Castilla y León, Universidad de Salamanca, Salamanca, Spain
- IBSAL, Salamanca, Spain
| | - Almudena Velasco
- Instituto de Neurociencias de Castilla y León, Universidad de Salamanca, Salamanca, Spain
- IBSAL, Salamanca, Spain
- * E-mail:
| |
Collapse
|
16
|
Dehghan S, Hesaraki M, Soleimani M, Mirnajafi-Zadeh J, Fathollahi Y, Javan M. Oct4 transcription factor in conjunction with valproic acid accelerates myelin repair in demyelinated optic chiasm in mice. Neuroscience 2016; 318:178-89. [PMID: 26804242 DOI: 10.1016/j.neuroscience.2016.01.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 12/23/2015] [Accepted: 01/13/2016] [Indexed: 12/26/2022]
Abstract
Multiple sclerosis is a demyelinating disease with severe neurological symptoms due to blockage of signal conduction in affected axons. Spontaneous remyelination via endogenous progenitors is limited and eventually fails. Recent reports showed that forced expression of some transcription factors within the brain converted somatic cells to neural progenitors and neuroblasts. Here, we report the effect of valproic acid (VPA) along with forced expression of Oct4 transcription factor on lysolecithin (LPC)-induced experimental demyelination. Mice were gavaged with VPA for one week, and then inducible Oct4 expressing lentiviral particles were injected into the lateral ventricle. After one-week induction of Oct4, LPC was injected into the optic chiasm. Functional remyelination was assessed by visual-evoked potential (VEP) recording. Myelination level was studied using FluoroMyelin staining and immunohistofluorescent (IHF) against proteolipid protein (PLP). IHF was also performed to detect Oct4 and SSEA1 as pluripotency markers and Olig2, Sox10, CNPase and PDGFRα as oligodendrocyte lineage markers. One week after injection of Oct4 expressing vector, pluripotency markers SSEA1 and Oct4 were detected in the rims of the 3rd ventricle. LPC injection caused extensive demyelination and significantly delayed the latency of VEP wave. Animals pre-treated with VPA+Oct4 expressing vector, showed faster recovery in the VEP latency and enhanced myelination. Immunostaining against oligodendrocyte lineage markers showed an increased number of Sox10+ and myelinating cells. Moreover, transdifferentiation of some Oct4-transfected cells (GFP+ cells) to Olig2+ and CNPase+ cells was confirmed by immunostaining. One-week administration of VPA followed by one-week forced expression of Oct4 enhanced myelination by converting transduced cells to myelinating oligodendrocytes. This finding seems promising for enhancing myelin repair within the adult brains.
Collapse
Affiliation(s)
- S Dehghan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - M Hesaraki
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - M Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - J Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Y Fathollahi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - M Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
17
|
Abstract
Oligodendrocyte precursor cells (OPCs) originate in the ventricular zones (VZs) of the brain and spinal cord and migrate throughout the developing central nervous system (CNS) before differentiating into myelinating oligodendrocytes (OLs). It is not known whether OPCs or OLs from different parts of the VZ are functionally distinct. OPCs persist in the postnatal CNS, where they continue to divide and generate myelinating OLs at a decreasing rate throughout adult life in rodents. Adult OPCs respond to injury or disease by accelerating their cell cycle and increasing production of OLs to replace lost myelin. They also form synapses with unmyelinated axons and respond to electrical activity in those axons by generating more OLs and myelin locally. This experience-dependent "adaptive" myelination is important in some forms of plasticity and learning, for example, motor learning. We review the control of OL lineage development, including OL population dynamics and adaptive myelination in the adult CNS.
Collapse
Affiliation(s)
- Dwight E Bergles
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, WBSB 1001, Baltimore, Maryland 21205
| | - William D Richardson
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
18
|
Sekizar S, Mannioui A, Azoyan L, Colin C, Thomas JL, Du Pasquier D, Mallat M, Zalc B. Remyelination by Resident Oligodendrocyte Precursor Cells in a Xenopus laevis Inducible Model of Demyelination. Dev Neurosci 2015; 37:232-42. [DOI: 10.1159/000380817] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 02/10/2015] [Indexed: 11/19/2022] Open
Abstract
We have generated a Xenopus laevis transgenic line, MBP-GFP-NTR, allowing conditional ablation of myelin-forming oligodendrocytes. In this transgenic line the transgene is driven by the proximal portion of the myelin basic protein regulatory sequence, specific to mature oligodendrocytes. The transgene protein is formed by the green fluorescent protein reporter fused to the Escherichia coli nitroreductase (NTR) selection enzyme. The NTR enzyme converts the innocuous prodrug metronidazole (MTZ) to a cytotoxin. Ablation of oligodendrocytes by MTZ treatment of the tadpole induced demyelination, and here we show that myelin debris are subsequently eliminated by microglial cells. After cessation of MTZ treatment, remyelination proceeded spontaneously. We questioned the origin of remyelinating cells. Our data suggest that Sox10+ oligodendrocyte precursor cells (OPCs), which are already present in the optic nerve prior to the experimentally induced demyelination, are responsible for remyelination, and this required only minimal (if any) cell division of OPCs.
Collapse
|
19
|
Pourabdolhossein F, Mozafari S, Morvan-Dubois G, Mirnajafi-Zadeh J, Lopez-Juarez A, Pierre-Simons J, Demeneix BA, Javan M. Nogo receptor inhibition enhances functional recovery following lysolecithin-induced demyelination in mouse optic chiasm. PLoS One 2014; 9:e106378. [PMID: 25184636 PMCID: PMC4153612 DOI: 10.1371/journal.pone.0106378] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Accepted: 07/29/2014] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Inhibitory factors have been implicated in the failure of remyelination in demyelinating diseases. Myelin associated inhibitors act through a common receptor called Nogo receptor (NgR) that plays critical inhibitory roles in CNS plasticity. Here we investigated the effects of abrogating NgR inhibition in a non-immune model of focal demyelination in adult mouse optic chiasm. METHODOLOGY/PRINCIPAL FINDINGS A focal area of demyelination was induced in adult mouse optic chiasm by microinjection of lysolecithin. To knock down NgR levels, siRNAs against NgR were intracerebroventricularly administered via a permanent cannula over 14 days, Functional changes were monitored by electrophysiological recording of latency of visual evoked potentials (VEPs). Histological analysis was carried out 3, 7 and 14 days post demyelination lesion. To assess the effect of NgR inhibition on precursor cell repopulation, BrdU was administered to the animals prior to the demyelination induction. Inhibition of NgR significantly restored VEPs responses following optic chiasm demyelination. These findings were confirmed histologically by myelin specific staining. siNgR application resulted in a smaller lesion size compared to control. NgR inhibition significantly increased the numbers of BrdU+/Olig2+ progenitor cells in the lesioned area and in the neurogenic zone of the third ventricle. These progenitor cells (Olig2+ or GFAP+) migrated away from this area as a function of time. CONCLUSIONS/SIGNIFICANCE Our results show that inhibition of NgR facilitate myelin repair in the demyelinated chiasm, with enhanced recruitment of proliferating cells to the lesion site. Thus, antagonizing NgR function could have therapeutic potential for demyelinating disorders such as Multiple Sclerosis.
Collapse
Affiliation(s)
- Fereshteh Pourabdolhossein
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Paris, France
| | - Sabah Mozafari
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ghislaine Morvan-Dubois
- UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Paris, France
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Alejandra Lopez-Juarez
- UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Paris, France
| | - Jacqueline Pierre-Simons
- UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Paris, France
| | - Barbara A. Demeneix
- UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Paris, France
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- * E-mail:
| |
Collapse
|
20
|
Mitew S, Hay C, Peckham H, Xiao J, Koenning M, Emery B. Mechanisms regulating the development of oligodendrocytes and central nervous system myelin. Neuroscience 2014; 276:29-47. [DOI: 10.1016/j.neuroscience.2013.11.029] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 11/13/2013] [Accepted: 11/14/2013] [Indexed: 12/29/2022]
|
21
|
de Castro F, Bribián A, Ortega MC. Regulation of oligodendrocyte precursor migration during development, in adulthood and in pathology. Cell Mol Life Sci 2013; 70:4355-68. [PMID: 23689590 PMCID: PMC11113994 DOI: 10.1007/s00018-013-1365-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 05/03/2013] [Accepted: 05/07/2013] [Indexed: 12/25/2022]
Abstract
Oligodendrocytes are the myelin-forming cells in the central nervous system (CNS). These cells originate from oligodendrocyte precursor cells (OPCs) during development, and they migrate extensively from oligodendrogliogenic niches along the neural tube to colonise the entire CNS. Like many other such events, this migratory process is precisely regulated by a battery of positional and signalling cues that act via their corresponding receptors and that are expressed dynamically by OPCs. Here, we will review the cellular and molecular basis of this important event during embryonic and postnatal development, and we will discuss the relevance of the substantial number of OPCs existing in the adult CNS. Similarly, we will consider the behaviour of OPCs in normal and pathological conditions, especially in animal models of demyelination and of the demyelinating disease, multiple sclerosis. The spontaneous remyelination observed after damage in demyelinating pathologies has a limited effect. Understanding the cellular and molecular mechanisms underlying the biology of OPCs, particularly adult OPCs, should help in the design of neuroregenerative strategies to combat multiple sclerosis and other demyelinating diseases.
Collapse
Affiliation(s)
- Fernando de Castro
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos-SESCAM, Finca "La Peraleda" s/n, 45071, Toledo, Spain,
| | | | | |
Collapse
|
22
|
Nagayama K, Kurita H, Nakamura M, Kusuda J, Tonari A, Takayama M, Fujioka Y, Shiokawa Y. Radiation-induced apoptosis of oligodendrocytes in the adult rat optic chiasm. Neurol Res 2013; 27:346-50. [PMID: 15949230 DOI: 10.1179/016164105x48833] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES The present study characterized glial cell injury provoked in adult rat chiasm within 24 hours after a single, high-dose irradiation of 20 Gy. METHODS All chiasmal glial cells in a section were counted, and the percentage of TUNEL-positive glial cells exhibiting apoptotic morphology was defined as the apoptotic rate. RESULTS Numbers of apoptotic cells increased significantly (p<0.0001) from 3 to 8 hours after exposure, but returned to baseline levels by 24 hours. Little evidence of apoptosis was observed in non-irradiated chiasms. Similar patterns of increase in apoptotic rate were observed in the genu of the corpus callosum, but the extent was significantly lower (p=0.047) in the optic chiasm, with a maximal rate of 1.9%. Immunohistochemically, apoptotic cells were positive for CNP, a marker for oligodendrocytes. DISCUSSION These data indicate that chiasmal irradiation induces limited, but significant apoptotic depletion of the oligodendroglial population, and may participate in the development of radiation-induced optic neuropathy.
Collapse
Affiliation(s)
- Kazuki Nagayama
- Department of Neurosurgery, Kyorin University School of Medicine, 6-20-2, Shinkawa, Mitaka-city, Tokyo 181-8611, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Solga AC, Gianino SM, Gutmann DH. NG2-cells are not the cell of origin for murine neurofibromatosis-1 (Nf1) optic glioma. Oncogene 2013; 33:289-99. [PMID: 23318450 DOI: 10.1038/onc.2012.580] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 10/26/2012] [Accepted: 10/26/2012] [Indexed: 01/19/2023]
Abstract
Low-grade glial neoplasms (astrocytomas) represent one of the most common brain tumors in the pediatric population. These tumors frequently form in the optic pathway (optic pathway gliomas, OPGs), especially in children with the neurofibromatosis type 1 (NF1)-inherited tumor predisposition syndrome. To model these tumors in mice, we have previously developed several Nf1 genetically-engineered mouse strains that form optic gliomas. However, there are three distinct macroglial cell populations in the optic nerve (astrocytes, NG2+ (nerve/glial antigen 2) cells and oligodendrocytes). The presence of NG2+ cells in the optic nerve raises the intriguing possibility that these cells could be the tumor-initiating cells, as has been suggested for adult glioma. In this report, we used a combination of complementary in vitro and novel genetically-engineered mouse strains in vivo to determine whether NG2+ cells could give rise to Nf1 optic glioma. First, we show that Nf1 inactivation results in a cell-autonomous increase in glial fibrillary acidic protein+ (GFAP+), but not in NG2+, cell proliferation in vitro. Second, similar to the GFAP-Cre transgenic strain that drives Nf1 optic gliomagenesis, NG2-expressing cells also give rise to all three macroglial lineages in vivo. Third, in contrast to the GFAP-Cre strain, Nf1 gene inactivation in NG2+ cells is not sufficient for optic gliomagenesis in vivo. Collectively, these data demonstrate that NG2+ cells are not the cell of origin for mouse optic glioma, and support a model in which gliomagenesis requires Nf1 loss in specific neuroglial progenitors during embryogenesis.
Collapse
Affiliation(s)
- A C Solga
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - S M Gianino
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - D H Gutmann
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
24
|
Sabo JK, Cate HS. Signalling pathways that inhibit the capacity of precursor cells for myelin repair. Int J Mol Sci 2013; 14:1031-49. [PMID: 23296277 PMCID: PMC3565305 DOI: 10.3390/ijms14011031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 12/21/2012] [Accepted: 12/31/2012] [Indexed: 12/17/2022] Open
Abstract
In demyelinating disorders such as Multiple Sclerosis (MS), targets of injury are myelin and oligodendrocytes, leading to severe neurological dysfunction. Regenerative therapies aimed at promoting oligodendrocyte maturation and remyelination are promising strategies for treatment in demyelinating disorders. Endogenous precursor cells or exogenous transplanted cells are potential sources for remyelinating oligodendrocytes in the central nervous system (CNS). Several signalling pathways have been implicated in regulating the capacity of these cell populations for myelin repair. Here, we review neural precursor cells and oligodendrocyte progenitor cells as potential sources for remyelinating oligodendrocytes and evidence for the functional role of key signalling pathways in inhibiting regeneration from these precursor cell populations.
Collapse
Affiliation(s)
- Jennifer K Sabo
- Centre for Neuroscience Research, Department of Anatomy and Neuroscience, University of Melbourne, Melbourne Brain Centre, Kenneth Myer Building, 30 Royal Parade, Parkville, Vic 3010, Australia.
| | | |
Collapse
|
25
|
Lee DY, Gianino SM, Gutmann DH. Innate neural stem cell heterogeneity determines the patterning of glioma formation in children. Cancer Cell 2012; 22:131-8. [PMID: 22789544 PMCID: PMC3396885 DOI: 10.1016/j.ccr.2012.05.036] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 04/07/2012] [Accepted: 05/31/2012] [Indexed: 11/19/2022]
Abstract
The concept that gliomas comprise a heterogeneous group of diseases distinguished by their developmental origin raises the intriguing possibility that neural stem cells (NSCs) from different germinal zones have differential capacities to respond to glioma-causing genetic changes. We demonstrate that lateral ventricle subventricular zone NSCs are molecularly and functionally distinct from those of the third ventricle. Consistent with a unique origin for pediatric low-grade glioma, third ventricle, but not lateral ventricle, NSCs hyperproliferate in response to mutations characteristic of childhood glioma. Finally, we demonstrate that pediatric optic gliomas in Nf1 genetically engineered mice arise from the third ventricle. Collectively, these observations establish the importance of innate brain region NSC heterogeneity in the patterning of gliomagenesis in children and adults.
Collapse
Affiliation(s)
- Da Yong Lee
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | |
Collapse
|
26
|
αB-Crystallin is Expressed in Myelinating Oligodendrocytes of the Developing and Adult Avian Retina. Neurochem Res 2012; 37:2135-42. [DOI: 10.1007/s11064-012-0835-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 06/14/2012] [Accepted: 06/23/2012] [Indexed: 10/28/2022]
|
27
|
Ortega MC, Cases O, Merchán P, Kozyraki R, Clemente D, de Castro F. Megalin mediates the influence of sonic hedgehog on oligodendrocyte precursor cell migration and proliferation during development. Glia 2012; 60:851-66. [PMID: 22354480 DOI: 10.1002/glia.22316] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 02/03/2012] [Indexed: 12/31/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) of the optic nerve are generated in the preoptic area, from where they migrate to colonize it entirely. Sonic hedgehog (Shh) induces the proliferation of these cells as well as influencing their migration, acting through its canonical receptor (Ptc-1). However, the multiligand receptor megalin (or LRP-2) is also involved in Shh-induced OPC proliferation and migration, and thus, we have evaluated the relevance of this interaction. During the stages at which Shh influences OPC development, we found megalin to be selectively expressed by optic nerve astrocytes, whereas Ptc-1 and Gli1 were found in OPCs. Indeed, this pattern of expression paralleled the rostral-caudal expression of the three Shh-related molecules during the time course of plp-dm20(+) -OPC colonization. The blockage of megalin partially abolished OPC chemoattraction and fully impaired Shh-induced proliferation. Using in vitro co-cultures of dissociated optic nerve cells, we demonstrated that Shh was internalized by astrocytes via megalin, and sufficient Shh was subsequently released to produce the biological effects on OPCs observed in the nerve. Together, these data indicate that at least part of the influence of Shh on OPCs is mediated by megalin during optic nerve development, and that astrocytes expressing megalin transiently capture Shh to present it to OPCs and/or to control the gradient of this molecule during development.
Collapse
Affiliation(s)
- María Cristina Ortega
- Grupo de Neurobiología del Desarrollo-GNDe, Unidad de Neurología Experimental, Hospital Nacional de Parapléjicos, Finca La Peraleda s/n, Toledo, Spain
| | | | | | | | | | | |
Collapse
|
28
|
Expression of Semaphorin 4F in neurons and brain oligodendrocytes and the regulation of oligodendrocyte precursor migration in the optic nerve. Mol Cell Neurosci 2012; 49:54-67. [PMID: 21945643 DOI: 10.1016/j.mcn.2011.09.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 09/09/2011] [Accepted: 09/12/2011] [Indexed: 11/22/2022] Open
Abstract
Semaphorins are secreted or membrane-anchored proteins that play critical roles in neural development and adult brain plasticity. Sema4F is a transmembrane semaphorin found on glutamatergic synapses, in which it is attached to the PSD-95-scaffolding protein. Here we further examined the expression of Sema4F by raising specific antibodies. We show that Sema4F protein is widely expressed by neurons during neural development and in the adult brain. We also demonstrate a preferential localization of this protein in postsynaptic dendrites. Moreover, Sema4F is expressed not only by neurons but also by oligodendrocyte precursors in the optic nerve and along the migratory pathways of oligodendroglial cells, and also by subsets of postnatal oligodendroglial cells in the brain. Finally, in vitro experiments demonstrate that endogenous Sema4F expressed by brain cells of oligodendroglial lineage regulates the outgrowth migration of oligodendrocyte precursors and promotes their differentiation. The present data extend our knowledge about the expression of Sema4F and uncover a novel function in the control of oligodendrocyte precursor migration in the developing brain.
Collapse
|
29
|
Gotoh H, Ueda T, Uno A, Ohuchi H, Ikenaka K, Ono K. Expression of myelin genes in the developing chick retina. Gene Expr Patterns 2011; 11:471-5. [PMID: 21872683 DOI: 10.1016/j.gep.2011.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 08/04/2011] [Accepted: 08/05/2011] [Indexed: 11/25/2022]
Abstract
In submammalian animals including chicks, the retina contains oligodendrocytes (OLs), and axons in the optic fiber layer are wrapped with compact myelin within the retina; however, the expression of myelin genes in the chick retina has not been demonstrated yet. In the present study, we examined the expression of three myelin genes (proteolipid protein, PLP; myelin basic protein, MBP; cyclic nucleotide phosphodiesterase, CNP) and PLP in the developing chick retina, in comparison to the localization of Mueller cells. In situ hybridization demonstrated that all three myelin genes began to be expressed at E14 in the chick embryo retina. They are mostly restricted to the ganglion cell layer and the optic fiber layer, with a few exceptions in the inner nuclear layer where Mueller cells reside; however, PLP mRNA+ cells do not express glutamine synthetase, or vice versa. The present results elucidate that myelin genes are expressed only by OLs that are mostly localized in the innermost layer of the developing chick retina.
Collapse
Affiliation(s)
- Hitosh Gotoh
- Department of Biology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | | | | | | | | |
Collapse
|
30
|
Analysis of Structural and Molecular Events Associated with Adult Rat Optic Chiasm and Nerves Demyelination and Remyelination; Possible Role for 3rd Ventricle Proliferating Cells. Neuromolecular Med 2011; 13:138-50. [DOI: 10.1007/s12017-011-8143-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Accepted: 01/05/2011] [Indexed: 12/31/2022]
|
31
|
Polster B, Crosier M, Lindsay S, Hayflick S. Expression of PLA2G6 in human fetal development: Implications for infantile neuroaxonal dystrophy. Brain Res Bull 2010; 83:374-9. [PMID: 20813170 PMCID: PMC2975838 DOI: 10.1016/j.brainresbull.2010.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Revised: 08/17/2010] [Accepted: 08/24/2010] [Indexed: 12/22/2022]
Abstract
Mutations in PLA2G6, which encodes calcium-independent phospholipase A(2) group VIA (iPLA2-VIA), underlie the autosomal recessive disorder infantile neuroaxonal dystrophy (INAD). INAD typically presents in the first year of life, and leads to optic atrophy and psychomotor regression. We have examined PLA2G6 expression in early human embryonic development by in situ hybridization. At Carnegie Stage (CS) 19 (approximately 7 post-conception weeks [PCW]), strong expression is evident in the ventricular zone (VZ) of midbrain and forebrain suggestive of expression in neural stem and progenitor cells. At CS23 (8PCW) expression is also detectable in the VZ of the hindbrain and the subventricular zone (SVZ) of the developing neocortex, ganglionic eminences and diencephalon. By 9PCW strong expression in the post-mitotic cells of the cortical plate can be seen in the developing neocortex. In the eye, expression is seen in the lens and retina at all stages examined. PLA2G6 expression is also evident in the alar plate of the spinal cord, dorsal root ganglia, the retina and lens in the eye and several non-neuronal tissues, including developing bones, lung, kidney and gut. These findings suggest a role for PLA2G6 in neuronal proliferation throughout the developing brain and in maturing neurons in the cortical plate and hindbrain. Although widespread PLA2G6 expression is detected in neuronal tissues, the pattern shows dynamic changes with time and indicates that INAD pathogenesis may begin prior to birth.
Collapse
Affiliation(s)
- Brenda Polster
- Molecular and Medical Genetics, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States.
| | | | | | | |
Collapse
|
32
|
Kim JY, Sohn HJ, Lee EY, Goo YS, Kim DW, Seo JH. Expression of αB-Crystallin in the Peripapillary Glial Cells of the Developing Chick Retina. Neurochem Res 2010; 36:76-82. [DOI: 10.1007/s11064-010-0266-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2010] [Indexed: 10/19/2022]
|
33
|
Garcia-Lopez R, Martinez S. Oligodendrocyte precursors originate in the parabasal band of the basal plate in prosomere 1 and migrate into the alar prosencephalon during chick development. Glia 2010; 58:1437-50. [DOI: 10.1002/glia.21019] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
34
|
Tsai HH, Macklin WB, Miller RH. Distinct modes of migration position oligodendrocyte precursors for localized cell division in the developing spinal cord. J Neurosci Res 2010; 87:3320-30. [PMID: 19301427 DOI: 10.1002/jnr.22058] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Establishment of the cytoarchitecture of the central nervous system reflects the stereotyped cell migration and proliferation of precursor cells during development. In vitro analyses have provided extensive information on the control of proliferation and differentiation of oligodendrocyte precursors (OPCs), but less is known about the migratory behavior of these cells in vivo. Here we utilize a transgenic mouse line expressing enhanced green fluorescent protein (EGFP) under the proteolipid protein promoter (PLP-EGFP mice) to visualize directly the behaviors of OPCs in developing spinal cord slices. During early development, OPCs disperse from their origin at the ventricular zone by using saltatory migration. This involves orientation of the cell with a leading edge toward the pial surface and alternating stationary and fast-moving phases and dramatic shape changes. Once cells exit the ventricular zone, they exhibit an exploratory mode of migration characterized by persistent translocation without dramatic changes in cell morphology. The control of migration, proliferation, and cytokinesis of OPCs appear to be closely linked. In netrin-1 mutant spinal cords that lack dispersal cues, OPC migration rates were not significantly different, but the trajectories were altered, and numbers of migrating cells were dramatically reduced. In contrast to DNA replication that occurs at the ventricular zone or throughout the spinal cord neuropil, cell division or cytokinesis of OPCs occurs predominantly at the interface between gray and white matters, with the majority of cleavage planes parallel to the pial surface. These studies suggest that positional cues are critical for regulating OPC behavior during spinal cord development.
Collapse
Affiliation(s)
- Hui-Hsin Tsai
- Center for Translational Neuroscience, Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | | | | |
Collapse
|
35
|
Abstract
Salla disease and infantile sialic acid storage disease are autosomal recessive lysosomal storage disorders caused by mutations in the gene encoding sialin, a membrane protein that transports free sialic acid out of the lysosome after it is cleaved from sialoglycoconjugates undergoing degradation. Accumulation of sialic acid in lysosomes defines these disorders, and the clinical phenotype is characterized by neurodevelopmental defects, including severe CNS hypomyelination. In this study, we used a sialin-deficient mouse to address how loss of sialin leads to the defect in myelination. Behavioral analysis of the sialin(-/-) mouse demonstrates poor coordination, seizures, and premature death. Analysis by histology, electron microscopy, and Western blotting reveals a decrease in myelination of the CNS but normal neuronal cytoarchitecture and normal myelination of the PNS. To investigate potential mechanisms underlying CNS hypomyelination, we studied myelination and oligodendrocyte development in optic nerves. We found reduced numbers of myelinated axons in optic nerves from sialin(-/-) mice, but the myelin that was present appeared grossly normal. Migration and density of oligodendrocyte precursor cells were normal; however, a marked decrease in the number of postmitotic oligodendrocytes and an associated increase in the number of apoptotic cells during the later stages of myelinogenesis were observed. These findings suggest that a defect in maturation of cells in the oligodendrocyte lineage leads to increased apoptosis and underlies the myelination defect associated with sialin loss.
Collapse
|
36
|
Bribián A, Esteban PF, Clemente D, Soussi-Yanicostas N, Thomas JL, Zalc B, de Castro F. A novel role for anosmin-1 in the adhesion and migration of oligodendrocyte precursors. Dev Neurobiol 2009; 68:1503-16. [PMID: 18814310 DOI: 10.1002/dneu.20678] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
At embryonic stages of development, oligodendrocyte precursors (OPCs) generated in the preoptic area colonize the entire optic nerve (ON). Different factors controlling migration of ON OPCs have been identified, including secreted growth factors, morphogens and guidance cues, as well as cell adhesion molecules. We have shown previously that the soluble form of the extracellular matrix (ECM) protein anosmin-1, impairs OPC migration induced by FGF-2. In the present work, we show that anosmin-1 is expressed by both migrating OPCs and axons of the retinal ganglion cells in the embryonic ON. In vitro, we observe that OPC migration is strongly impaired by contact with anosmin-1 when used as a substrate and, in contrast to previous results, this effect is independent of FGF-2/FGFR1 signaling. We also show that OPCs preferentially adhere to anosmin-1 when compared with other ECM molecules used as substrates, and that when the endogenous anosmin-1 expressed by OPCs is blocked, OPC adhesion to all the different substrates (including anosmin-1), is significantly reduced. This novel effect of anosmin-1 on cell adhesion is also independent of FGF-2/FGFR1. We finally demonstrate that the blockade of the endogenous anosmin-1 expressed by OPCs impairs their migration. Our data suggest that the endogenous anosmin-1 expressed by OPCs is necessary for the correct adhesion of these cells to the different components of the ECM (including anosmin-1 itself), contributing to the migration of these cells.
Collapse
Affiliation(s)
- Ana Bribián
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca La Peraleda s/n, Toledo E-45071, Spain
| | | | | | | | | | | | | |
Collapse
|
37
|
Structural requirement of TAG-1 for retinal ganglion cell axons and myelin in the mouse optic nerve. J Neurosci 2008; 28:7624-36. [PMID: 18650339 DOI: 10.1523/jneurosci.1103-08.2008] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
White matter axons organize into fascicles that grow over long distances and traverse very diverse environments. The molecular mechanisms preserving this structure of white matter axonal tracts are not well known. Here, we used the optic nerve as a model and investigated the role of TAG-1, a cell adhesion molecule expressed by retinal axons. TAG-1 was first expressed in the embryonic retinal ganglion cells (RGCs) and later in the postnatal myelin-forming cells in the optic nerve. We describe the consequences of genetic loss of Tag-1 on the developing and adult retinogeniculate tract. Tag-1-null embryos display anomalies in the caliber of RGC axons, associated with an abnormal organization of the astroglial network in the optic nerve. The contralateral projections in the lateral geniculate nucleus are expanded postnatally. In the adult, Tag-1-null mice show a loss of RGC axons, with persistent abnormalities of axonal caliber and additional cytoskeleton and myelination defects. Therefore, TAG-1 is an essential regulator of the structure of RGC axons and their surrounding glial cells in the optic nerve.
Collapse
|
38
|
Expression of transferrin binding protein in the capillaries of the brain in the developing chick embryo. Neurochem Res 2008; 33:2288-93. [PMID: 18459044 DOI: 10.1007/s11064-008-9716-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2008] [Accepted: 04/10/2008] [Indexed: 10/22/2022]
Abstract
Transferrin-binding protein (TfBP) has been shown to be a novel protein, structurally related to the chicken heat shock protein 108. The physiological function of this protein, however, has not yet been established. Antiserum to TfBP selectively stains transferrin- and iron-rich oligodendrocytes and choroidal epithelium in the adult and embryonic chick brain, suggesting a role for this protein in transferrin and iron storage in these cells. In this study, we further demonstrate TfBP-immunoreactivity (IR) in the blood vessels of the embryonic chick central nervous system. A strong TfBP-IR was present in blood vessels from E6, declined from E10 and was absent by E18. Thus, the expression of the TfBP in the blood vessels precedes its expression in the oligodendrocytes. At the subcellular level, TfBP-IR was confined to the cytoplasm of capillary pericytes while the Tf-receptor IR was associated with the capillary endothelium of the brain. The up-regulated expression of TfBP, together with the Tf-receptor of the brain capillaries, suggests that pericytes may be associated with the high iron uptake required for the metabolic demands of the developing brain.
Collapse
|
39
|
Spatiotemporal Gradient of Astrocyte Development in the Chick Optic Tectum: Evidence for Multiple Origins and Migratory Paths of Astrocytes. Neurochem Res 2008; 33:1346-55. [DOI: 10.1007/s11064-008-9590-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2007] [Accepted: 01/02/2008] [Indexed: 10/22/2022]
|
40
|
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
|
41
|
Baracskay KL, Kidd GJ, Miller RH, Trapp BD. NG2-positive cells generate A2B5-positive oligodendrocyte precursor cells. Glia 2007; 55:1001-10. [PMID: 17503442 DOI: 10.1002/glia.20519] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cellular specification of the oligodendrocyte lineage occurs through a series of stages identified by expression of distinct biochemical characteristics. The best characterized oligodendrocyte progenitor cell (OPC) in vitro is the bipotential O2-A progenitor, identified by labeling with monoclonal antibody A2B5, which proliferates predominantly in response to platelet derived growth factor (PDGF). The cellular ancestors of O2-A progenitor cells are currently unclear. In vivo OPCs can be identified by expression of the cell surface markers NG2 (a sulfated proteoglycan) and platelet derived growth factor receptor alphaR). Substantial evidence supports the generation of oligodendrocytes from NG2(+), PDGFalphaR(+) cells both in vivo and in vitro. The developmental relationship between NG2(+) cells and A2B5(-) positive cells is unknown and it is unclear whether they represent identical, partially overlapping or nonoverlapping populations of cells. Here we show that in cultures of developing brain NG2(+) and A2B5(+) cells arise from overlapping cell populations. NG2(+) cells appear prior to the expression of A2B5(+) cells and generate A2B5(+) cells. We propose that during development NG2(+)/A2B5(-) cells (pre-OPCs) represent the direct ancestor to A2B5(+) O2A progenitor cells (OPCs).
Collapse
Affiliation(s)
- Karen L Baracskay
- Department of Neuroscience, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | | | | | | |
Collapse
|
42
|
Merchán P, Bribián A, Sánchez-Camacho C, Lezameta M, Bovolenta P, de Castro F. Sonic hedgehog promotes the migration and proliferation of optic nerve oligodendrocyte precursors. Mol Cell Neurosci 2007; 36:355-68. [PMID: 17826177 DOI: 10.1016/j.mcn.2007.07.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2007] [Revised: 07/19/2007] [Accepted: 07/24/2007] [Indexed: 11/26/2022] Open
Abstract
Optic nerve (ON) oligodendrocyte precursors (OPCs) are generated under the influence of the Sonic hedgehog (Shh) in the preoptic area from where they migrate to colonise the entire nerve. The molecular events that control this migration are still poorly understood. Recent studies suggested that Shh is often used by the same cell population to control different processes, including cell proliferation and migration, raising the possibility that Shh could contribute to these aspects of OPC development. In support of this idea, we show here that Shh induces the proliferation of OPCs derived from embryonic mouse ON explants and acts as a chemoattractant for their migration. In ovo injections of hybridomas secreting Shh-specific blocking antibody decreases the number of OPCs present in chick ONs, particularly in the retinal portion of the nerve. Altogether these data indicate that Shh contributes to OPC proliferation and distribution along the ON, in addition to their specification.
Collapse
Affiliation(s)
- Paloma Merchán
- Grupo de Neurobiología del Desarrollo, Hospital Nacional de Parapléjicos, Finca La Peraleda, s/n, E-45071-Toledo, Spain
| | | | | | | | | | | |
Collapse
|
43
|
Benowitz L, Yin Y. Rewiring the injured CNS: lessons from the optic nerve. Exp Neurol 2007; 209:389-98. [PMID: 17610877 PMCID: PMC2323976 DOI: 10.1016/j.expneurol.2007.05.025] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2007] [Accepted: 05/22/2007] [Indexed: 11/30/2022]
Abstract
The optic nerve offers a number of advantages for investigating mechanisms that govern axon regeneration in the CNS. Although mature retinal ganglion cells (RGCs) normally show no ability to regenerate injured axons through the optic nerve, this situation can be partially reversed by inducing an inflammatory response in the eye. The secretion of a previously unknown growth factor, oncomodulin, along with co-factors, causes RGCs to undergo dramatic changes in gene expression and regenerate lengthy axons into the highly myelinated optic nerve. By themselves, strategies that counteract inhibitory signals associated with myelin and the glial scar are insufficient to promote extensive regeneration in this system. However, combinatorial treatments that activate neurons' intrinsic growth state and overcome inhibitory signals result in dramatic axon regeneration in vivo. Because of the ease of introducing trophic factors, soluble receptors, drugs, or viruses expressing any gene or small interfering RNA of interest into RGCs, this system is ideal for identifying intracellular signaling pathways, transcriptional cascades, and ligand-receptor interactions that enable axon regeneration to occur in the CNS.
Collapse
Affiliation(s)
- Larry Benowitz
- Laboratories for Neuroscience Research in Neurosurgery and Neurobiology Program, Children's Hospital, Boston, MA 02115, USA.
| | | |
Collapse
|
44
|
Freret ME, Gutmann DH. Optic pathway gliomas in neurofibromatosis-1: controversies and recommendations. Ann Neurol 2007; 61:189-98. [PMID: 17387725 PMCID: PMC5908242 DOI: 10.1002/ana.21107] [Citation(s) in RCA: 367] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Optic pathway glioma (OPG), seen in 15% to 20% of individuals with neurofibromatosis type 1 (NF1), account for significant morbidity in young children with NF1. Overwhelmingly a tumor of children younger than 7 years, OPG may present in individuals with NF1 at any age. Although many OPG may remain indolent and never cause signs or symptoms, others lead to vision loss, proptosis, or precocious puberty. Because the natural history and treatment of NF1-associated OPG is different from that of sporadic OPG in individuals without NF1, a task force composed of basic scientists and clinical researchers was assembled in 1997 to propose a set of guidelines for the diagnosis and management of NF1-associated OPG. This new review highlights advances in our understanding of the pathophysiology and clinical behavior of these tumors made over the last 10 years. Controversies in both the diagnosis and management of these tumors are examined. Finally, specific evidence-based recommendations are proposed for clinicians caring for children with NF1.
Collapse
Affiliation(s)
| | - David H. Gutmann
- To whom correspondence should be addressed: Telephone: 314-362-7379, Facsimile: 314-362-2388,
| |
Collapse
|
45
|
Harada T, Harada C, Parada LF. Molecular regulation of visual system development: more than meets the eye. Genes Dev 2007; 21:367-78. [PMID: 17322396 DOI: 10.1101/gad.1504307] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Vertebrate eye development has been an excellent model system to investigate basic concepts of developmental biology ranging from mechanisms of tissue induction to the complex patterning and bidimensional orientation of the highly specialized retina. Recent advances have shed light on the interplay between numerous transcriptional networks and growth factors that are involved in the specific stages of retinogenesis, optic nerve formation, and topographic mapping. In this review, we summarize this recent progress on the molecular mechanisms underlying the development of the eye, visual system, and embryonic tumors that arise in the optic system.
Collapse
Affiliation(s)
- Takayuki Harada
- Department of Developmental Biology, Kent Waldrep Foundation Center for Basic Neuroscience Research on Nerve Growth and Regeneration, University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | | | | |
Collapse
|
46
|
Soukkarieh C, Agius E, Soula C, Cochard P. Pax2 regulates neuronal-glial cell fate choice in the embryonic optic nerve. Dev Biol 2006; 303:800-13. [PMID: 17173889 DOI: 10.1016/j.ydbio.2006.11.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2006] [Revised: 11/06/2006] [Accepted: 11/07/2006] [Indexed: 01/04/2023]
Abstract
During development, neural cell fate in the vertebrate optic nerve is restricted to the astroglial lineage. However, when isolated from the embryo and explanted in vitro, optic nerve progenitors generate neurons instead of astrocytes, suggesting that neuronal potentialities exist and are repressed in progenitors in vivo. Here we have investigated the mechanisms controlling cell fate in the optic nerve. The optic nerve is characterized by expression of the homeodomain transcription factor Pax2 which is maintained in differentiated astrocytes. We have observed that Pax2 is rapidly down-regulated in explanted optic nerves that generate neurons, and that its overexpression by electroporation in the optic nerve, or ectopically in the neural tube, is sufficient to block neuronal differentiation and allow glial development, showing that Pax2 plays a major role in controlling cell fate in the optic nerve. In vitro and ex vivo experiments further show that a signaling cascade that involves successively Sonic hedgehog and FGF is required to maintain Pax2 expression in optic nerve precursors whereby inhibiting the neuronal fate and promoting astroglial differentiation.
Collapse
Affiliation(s)
- Chadi Soukkarieh
- Centre de Biologie du Développement, CNRS UMR 5547, Institut d'Exploration Fonctionnelle des Génomes, Université Paul Sabatier, 118 Route de Narbonne, 31062 Toulouse Cedex, France
| | | | | | | |
Collapse
|
47
|
Bonfanti L. PSA-NCAM in mammalian structural plasticity and neurogenesis. Prog Neurobiol 2006; 80:129-64. [PMID: 17029752 DOI: 10.1016/j.pneurobio.2006.08.003] [Citation(s) in RCA: 339] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2006] [Revised: 08/04/2006] [Accepted: 08/21/2006] [Indexed: 12/14/2022]
Abstract
Polysialic acid (PSA) is a linear homopolymer of alpha2-8-N acetylneuraminic acid whose major carrier in vertebrates is the neural cell adhesion molecule (NCAM). PSA serves as a potent negative regulator of cell interactions via its unusual biophysical properties. PSA on NCAM is developmentally regulated thus playing a prominent role in different forms of neural plasticity spanning from embryonic to adult nervous system, including axonal growth, outgrowth and fasciculation, cell migration, synaptic plasticity, activity-induced plasticity, neuronal-glial plasticity, embryonic and adult neurogenesis. The cellular distribution, developmental changes and possible function(s) of PSA-NCAM in the central nervous system of mammals here are reviewed, along with recent findings and theories about the relationships between NCAM protein and PSA as well as the role of different polysialyltransferases. Particular attention is focused on postnatal/adult neurogenesis, an issue which has been deeply investigated in the last decade as an example of persisting structural plasticity with potential implications for brain repair strategies. Adult neurogenic sites, although harbouring all subsequent steps of cell differentiation, from stem cell division to cell replacement, do not faithfully recapitulate development. After birth, they undergo morphological and molecular modifications allowing structural plasticity to adapt to the non-permissive environment of the mature nervous tissue, that are paralled by changes in the expression of PSA-NCAM. The use of PSA-NCAM as a marker for exploring differences in structural plasticity and neurogenesis among mammalian species is also discussed.
Collapse
Affiliation(s)
- Luca Bonfanti
- Department of Veterinary Morphophysiology, University of Turin, Via Leonardo da Vinci 44, 10095 Grugliasco, Italy.
| |
Collapse
|
48
|
Gao L, Miller RH. Specification of optic nerve oligodendrocyte precursors by retinal ganglion cell axons. J Neurosci 2006; 26:7619-28. [PMID: 16855089 PMCID: PMC6674293 DOI: 10.1523/jneurosci.0855-06.2006] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Cell fate commitment in the developing CNS frequently depends on localized cell-cell interactions. In the avian visual system the optic nerve oligodendrocytes are derived from founder cells located at the floor of the third ventricle. Here we show that the induction of these founder cells is directly dependent on signaling from the retinal ganglion cell (RGC) axons. The appearance of oligodendrocyte precursor cells (OPCs) correlates with the projection of RGC axons, and early eye removal dramatically reduces the number of OPCs. In vitro signaling from retinal neurites induces OPCs in responsive tissue. Retinal axon induction of OPCs is dependent on sonic hedgehog (Shh) and neuregulin signaling, and the inhibition of either signal reduces OPC induction in vivo and in vitro. The dependence of OPCs on retinal axonal cues appears to be a common phenomenon, because ocular retardation (or(J)) mice lacking optic nerve have dramatically reduced OPCs in the midline of the third ventricle.
Collapse
|
49
|
Bribián A, Barallobre MJ, Soussi-Yanicostas N, de Castro F. Anosmin-1 modulates the FGF-2-dependent migration of oligodendrocyte precursors in the developing optic nerve. Mol Cell Neurosci 2006; 33:2-14. [PMID: 16876430 DOI: 10.1016/j.mcn.2006.05.009] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2006] [Revised: 05/11/2006] [Accepted: 05/19/2006] [Indexed: 11/29/2022] Open
Abstract
Oligodendrocyte precursors (OPCs) originate at specific domains within the neural tube before migrating to colonize the entire CNS. Once in their target areas, these cells differentiate into oligodendrocytes, the myelin-forming cells in the CNS. Using the embryonic mouse optic nerve as an experimental model, we have analyzed the influence of FGF-2 on OPC development. FGF-2 exerts a dose-dependent motogenic effect on the migration of plp-dm20+ and it also acts as a chemoattractant on these cells. These effects produced by FGF-2 are principally mediated by the FGFR1 receptor, which is expressed by OPCs. Anosmin-1 is the protein that is defective in the X-linked form of human Kallmann syndrome. This protein is expressed by retinal axons and it also interacts with FGFR1, thereby impairing the migration of OPCs. Because both Anosmin-1 and FGF-2 are present in the optic nerve in vivo, we propose a model whereby the relative concentration of these two proteins modulates the migration of OPCs during development through their interaction with FGFR1. This FGF-2/FGFR1/Anosmin-1 system may be relevant in the context of demyelinating diseases.
Collapse
Affiliation(s)
- Ana Bribián
- Instituto de Neurociencias de Castilla y León-INCyL, Universidad de Salamanca, Avda. de Alfonso X el Sabio, s/n, E-37007-Salamanca, Spain
| | | | | | | |
Collapse
|
50
|
Abstract
Olig2 is a basic helix-loop-helix transcription factor that is specifically expressed in oligodendrocyte precursors in the central nervous system. Owing to the absence of oligodendrocytes in the retina, however, the role of olig2 in retinal development remains unclear. In the present study, we demonstrated for the first time that olig2 is expressed in embryonic and postnatal mouse retina. Interestingly, a subset of the bromodeoxyuridine-positive cells expressed olig2 in the embryonic retina. On the other hand, olig2-positive cells did not express nestin, neuron-specific class III beta-tubulin, neurofilament, or various retinal cell-type-specific markers. These results suggest that olig2 is involved in the maintenance of the undifferentiated state of retinal progenitor cells.
Collapse
Affiliation(s)
- Kazuaki Nakamura
- Department of Molecular Neurobiology, Tokyo Metropolitan Institute for Neuroscience, Fuchu, Tokyo , Japan
| | | | | | | |
Collapse
|