1
|
Soares LC, Al-Dalahmah O, Hillis J, Young CC, Asbed I, Sakaguchi M, O’Neill E, Szele FG. Novel Galectin-3 Roles in Neurogenesis, Inflammation and Neurological Diseases. Cells 2021; 10:3047. [PMID: 34831271 PMCID: PMC8618878 DOI: 10.3390/cells10113047] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/16/2022] Open
Abstract
Galectin-3 (Gal-3) is an evolutionarily conserved and multifunctional protein that drives inflammation in disease. Gal-3's role in the central nervous system has been less studied than in the immune system. However, recent studies show it exacerbates Alzheimer's disease and is upregulated in a large variety of brain injuries, while loss of Gal-3 function can diminish symptoms of neurodegenerative diseases such as Alzheimer's. Several novel molecular pathways for Gal-3 were recently uncovered. It is a natural ligand for TREM2 (triggering receptor expressed on myeloid cells), TLR4 (Toll-like receptor 4), and IR (insulin receptor). Gal-3 regulates a number of pathways including stimulation of bone morphogenetic protein (BMP) signaling and modulating Wnt signalling in a context-dependent manner. Gal-3 typically acts in pathology but is now known to affect subventricular zone (SVZ) neurogenesis and gliogenesis in the healthy brain. Despite its myriad interactors, Gal-3 has surprisingly specific and important functions in regulating SVZ neurogenesis in disease. Gal-1, a similar lectin often co-expressed with Gal-3, also has profound effects on brain pathology and adult neurogenesis. Remarkably, Gal-3's carbohydrate recognition domain bears structural similarity to the SARS-CoV-2 virus spike protein necessary for cell entry. Gal-3 can be targeted pharmacologically and is a valid target for several diseases involving brain inflammation. The wealth of molecular pathways now known further suggest its modulation could be therapeutically useful.
Collapse
Affiliation(s)
- Luana C. Soares
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford OX1 3QX, UK; (L.C.S.); (I.A.)
- Department of Oncology, University of Oxford, Oxford OX1 3QX, UK;
| | - Osama Al-Dalahmah
- Irving Medical Center, Columbia University, New York, NY 10032, USA;
| | - James Hillis
- Massachusets General Hospital, Harvard Medical School, 15 Parkman Street, Boston, MA 02114, USA;
| | - Christopher C. Young
- Department of Neurological Surgery, University of Washington, 325 Ninth Avenue, Seattle, WA 98104, USA;
| | - Isaiah Asbed
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford OX1 3QX, UK; (L.C.S.); (I.A.)
| | - Masanori Sakaguchi
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba 305-8575, Japan;
| | - Eric O’Neill
- Department of Oncology, University of Oxford, Oxford OX1 3QX, UK;
| | - Francis G. Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford OX1 3QX, UK; (L.C.S.); (I.A.)
| |
Collapse
|
2
|
Mecha M, Yanguas-Casás N, Feliú A, Mestre L, Carrillo-Salinas FJ, Riecken K, Gomez-Nicola D, Guaza C. Involvement of Wnt7a in the role of M2c microglia in neural stem cell oligodendrogenesis. J Neuroinflammation 2020; 17:88. [PMID: 32192522 PMCID: PMC7081569 DOI: 10.1186/s12974-020-01734-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 02/03/2020] [Indexed: 12/22/2022] Open
Abstract
Background The participation of microglia in CNS development and homeostasis indicate that these cells are pivotal for the regeneration that occurs after demyelination. The clearance of myelin debris and the inflammatory-dependent activation of local oligodendrocyte progenitor cells in a demyelinated lesion is dependent on the activation of M2c microglia, which display both phagocytic and healing functions. Emerging interest has been raised about the role of Wnt/β-catenin signaling in oligodendrogenesis and myelination. Besides, cytokines and growth factors released by microglia can control the survival, proliferation, migration, and differentiation of neural stem cells (NSCs), contributing to remyelination through the oligodendrocyte specification of this adult neurogenic niche. Methods TMEV-IDD model was used to study the contribution of dorsal SVZ stem cells to newly born oligodendrocytes in the corpus callosum following demyelination by (i) en-face dorsal SVZ preparations; (ii) immunohistochemistry; and (iii) cellular tracking. By RT-PCR, we analyzed the expression of Wnt proteins in demyelinated and remyelinating corpus callosum. Using in vitro approaches with microglia cultures and embryonic NSCs, we studied the role of purified myelin, Wnt proteins, and polarized microglia-conditioned medium to NSC proliferation and differentiation. One-way ANOVA followed by Bonferroni’s post-hoc test, or a Student’s t test were used to establish statistical significance. Results The demyelination caused by TMEV infection is paralleled by an increase in B1 cells and pinwheels in the dorsal SVZ, resulting in the mobilization of SVZ proliferative progenitors and their differentiation into mature oligodendrocytes. Demyelination decreased the gene expression of Wnt5a and Wnt7a, which was restored during remyelination. In vitro approaches show that Wnt3a enhances NSC proliferation, while Wnt7a and myelin debris promotes oligodendrogenesis from NSCs. As phagocytic M2c microglia secrete Wnt 7a, their conditioned media was found to induce Wnt/β-Catenin signaling in NSCs promoting an oligodendroglial fate. Conclusions We define here the contribution of microglia to Wnt production depending on their activation state, with M1 microglia secreting the Wnt5a protein and M2c microglia secreting Wnt7a. Collectively, our data reveal the role of reparative microglia in NSC oligodendrogenesis with the involvement of Wnt7a.
Collapse
Affiliation(s)
- Miriam Mecha
- Departamento de Neurobiología Funcional y de Sistemas, Grupo de Neuroinmunología, Instituto Cajal, CSIC, Madrid, Spain.
| | - Natalia Yanguas-Casás
- Departamento de Neurobiología Funcional y de Sistemas, Grupo de Neuroinmunología, Instituto Cajal, CSIC, Madrid, Spain.,Present address: Grupo de Investigación en Linfomas, Instituto Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Majadahonda, Madrid, Spain
| | - Ana Feliú
- Departamento de Neurobiología Funcional y de Sistemas, Grupo de Neuroinmunología, Instituto Cajal, CSIC, Madrid, Spain
| | - Leyre Mestre
- Departamento de Neurobiología Funcional y de Sistemas, Grupo de Neuroinmunología, Instituto Cajal, CSIC, Madrid, Spain
| | | | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Clinic for Stem Cell Transplantation, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Diego Gomez-Nicola
- Centre for Biological Sciences, University of Southampton, Southampton, UK
| | - Carmen Guaza
- Departamento de Neurobiología Funcional y de Sistemas, Grupo de Neuroinmunología, Instituto Cajal, CSIC, Madrid, Spain.
| |
Collapse
|
3
|
Bardella C, Al-Shammari AR, Soares L, Tomlinson I, O'Neill E, Szele FG. The role of inflammation in subventricular zone cancer. Prog Neurobiol 2018; 170:37-52. [PMID: 29654835 DOI: 10.1016/j.pneurobio.2018.04.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 03/10/2018] [Accepted: 04/07/2018] [Indexed: 12/12/2022]
Abstract
The adult subventricular zone (SVZ) stem cell niche has proven vital for discovering neurodevelopmental mechanisms and holds great potential in medicine for neurodegenerative diseases. Yet the SVZ holds a dark side - it can become tumorigenic. Glioblastomas can arise from the SVZ via cancer stem cells (CSCs). Glioblastoma and other brain cancers often have dismal prognoses since they are resistant to treatment. In this review we argue that the SVZ is susceptible to cancer because it contains stem cells, migratory progenitors and unusual inflammation. Theoretically, SVZ stem cells can convert to CSCs more readily than can postmitotic neural cells. Additionally, the robust long-distance migration of SVZ progenitors can be subverted upon tumorigenesis to an infiltrative phenotype. There is evidence that the SVZ, even in health, exhibits chronic low-grade cellular and molecular inflammation. Its inflammatory response to brain injuries and disease differs from that of other brain regions. We hypothesize that the SVZ inflammatory environment can predispose cells to novel mutations and exacerbate cancer phenotypes. This can be studied in animal models in which human mutations related to cancer are knocked into the SVZ to induce tumorigenesis and the CSC immune interactions that precede full-blown cancer. Importantly inflammation can be pharmacologically modulated providing an avenue to brain cancer management and treatment. The SVZ is accessible by virtue of its location surrounding the lateral ventricles and CSCs in the SVZ can be targeted with a variety of pharmacotherapies. Thus, the SVZ can yield aggressive tumors but can be targeted via several strategies.
Collapse
Affiliation(s)
- Chiara Bardella
- Institute of Cancer and Genomics Sciences, University of Birmingham, Birmingham, UK
| | - Abeer R Al-Shammari
- Research and Development, Qatar Research Leadership Program, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Luana Soares
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Department of Oncology, University of Oxford, Oxford, UK
| | - Ian Tomlinson
- Institute of Cancer and Genomics Sciences, University of Birmingham, Birmingham, UK
| | - Eric O'Neill
- Department of Oncology, University of Oxford, Oxford, UK
| | - Francis G Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
4
|
Laso-García F, Ramos-Cejudo J, Carrillo-Salinas FJ, Otero-Ortega L, Feliú A, Gómez-de Frutos M, Mecha M, Díez-Tejedor E, Guaza C, Gutiérrez-Fernández M. Therapeutic potential of extracellular vesicles derived from human mesenchymal stem cells in a model of progressive multiple sclerosis. PLoS One 2018; 13:e0202590. [PMID: 30231069 PMCID: PMC6145506 DOI: 10.1371/journal.pone.0202590] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 08/05/2018] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) have emerged as important mediators of intercellular communication and as possible therapeutic agents in inflammation-mediated demyelinating diseases, including multiple sclerosis (MS). In the present study, we investigated whether intravenously administered EVs derived from mesenchymal stem cells (MSCs) from human adipose tissue might mediate recovery in Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease, a progressive model of MS. SJL/J mice were subjected to EV treatment once the disease was established. We found that intravenous EV administration improved motor deficits, reduced brain atrophy, increased cell proliferation in the subventricular zone and decreased inflammatory infiltrates in the spinal cord in mice infected with TMEV. EV treatment was also capable of modulating neuroinflammation, given glial fibrillary acidic protein and Iba-1 staining were reduced in the brain, whereas myelin protein expression was increased. Changes in the morphology of microglial cells in the spinal cord suggest that EVs also modulate the activation state of microglia. The clear reduction in plasma cytokine levels, mainly in the Th1 and Th17 phenotypes, in TMEV mice treated with EVs confirms the immunomodulatory ability of intravenous EVs. According to our results, EV administration attenuates motor deficits through immunomodulatory actions, diminishing brain atrophy and promoting remyelination. Further studies are necessary to establish EV delivery as a possible therapy for the neurodegenerative phase of MS.
Collapse
Affiliation(s)
- Fernando Laso-García
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Madrid, Spain
| | - Jaime Ramos-Cejudo
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Madrid, Spain
| | | | - Laura Otero-Ortega
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Madrid, Spain
| | - Ana Feliú
- Functional and Systems Neurobiology Department, Neuroimmunology Group, Cajal Institute, Madrid, Spain
| | - MariCarmen Gómez-de Frutos
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Madrid, Spain
| | - Miriam Mecha
- Functional and Systems Neurobiology Department, Neuroimmunology Group, Cajal Institute, Madrid, Spain
| | - Exuperio Díez-Tejedor
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Madrid, Spain
| | - Carmen Guaza
- Functional and Systems Neurobiology Department, Neuroimmunology Group, Cajal Institute, Madrid, Spain
| | - María Gutiérrez-Fernández
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Madrid, Spain
| |
Collapse
|
5
|
El Waly B, Cayre M, Durbec P. Promoting Myelin Repair through In Vivo Neuroblast Reprogramming. Stem Cell Reports 2018; 10:1492-1504. [PMID: 29606615 PMCID: PMC5995160 DOI: 10.1016/j.stemcr.2018.02.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/26/2018] [Accepted: 02/28/2018] [Indexed: 11/28/2022] Open
Abstract
Demyelination is frequently observed in a variety of CNS insults and neurodegenerative diseases. In rodents, adult neural stem cells can generate oligodendrocytes and participate to myelin repair. However, these cells mainly produce migratory neuroblasts that differentiate in the olfactory bulb. Here, we show that, in the demyelination context, a small subset of these neuroblasts can spontaneously convert into myelinating oligodendrocytes. Furthermore, we demonstrate that the contribution of neuroblasts to myelin repair can be improved by in vivo forced expression of two transcription factors: OLIG2 and SOX10. These factors promote directed fate conversion of endogenous subventricular zone neuroblasts into mature functional oligodendrocytes, leading to enhanced remyelination in a cuprizone-induced mouse model of demyelination. These findings highlight the unexpected plasticity of committed neuroblasts and provide proof of concept that they could be targeted for the treatment of demyelinated lesions in the adult brain. Sox10 and Olig2 convert endogenous neuroblasts into myelinating oligodendrocytes Converted cells migrate to the corpus callosum, striatum, and cortex Converted cells produce myelin and participate to the formation of node of Ranvier Forced neuroblast conversion improves myelin regeneration
Collapse
Affiliation(s)
- Bilal El Waly
- 1-Aix Marseille University, CNRS, IBDM-UMR 7288, Case 907, Parc Scientifique de Luminy, campus de Luminy, 13288 Marseille, Cedex 09, France
| | - Myriam Cayre
- 1-Aix Marseille University, CNRS, IBDM-UMR 7288, Case 907, Parc Scientifique de Luminy, campus de Luminy, 13288 Marseille, Cedex 09, France
| | - Pascale Durbec
- 1-Aix Marseille University, CNRS, IBDM-UMR 7288, Case 907, Parc Scientifique de Luminy, campus de Luminy, 13288 Marseille, Cedex 09, France.
| |
Collapse
|
6
|
Adorjan I, Ahmed B, Feher V, Torso M, Krug K, Esiri M, Chance SA, Szele FG. Calretinin interneuron density in the caudate nucleus is lower in autism spectrum disorder. Brain 2017; 140:2028-2040. [PMID: 29177493 DOI: 10.1093/brain/awx131] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 04/18/2017] [Indexed: 02/05/2023] Open
Abstract
Autism spectrum disorder is a debilitating condition with possible neurodevelopmental origins but unknown neuroanatomical correlates. Whereas investigators have paid much attention to the cerebral cortex, few studies have detailed the basal ganglia in autism. The caudate nucleus may be involved in the repetitive movements and limbic changes of autism. We used immunohistochemistry for calretinin and neuropeptide Y in 24 age- and gender-matched patients with autism spectrum disorder and control subjects ranging in age from 13 to 69 years. Patients with autism had a 35% lower density of calretinin+ interneurons in the caudate that was driven by loss of small calretinin+ neurons. This was not caused by altered size of the caudate, as its cross-sectional surface areas were similar between diagnostic groups. Controls exhibited an age-dependent increase in the density of medium and large calretinin+ neurons, whereas subjects with autism did not. Diagnostic groups did not differ regarding ionized calcium-binding adapter molecule 1+ immunoreactivity for microglia, suggesting chronic inflammation did not cause the decreased calretinin+ density. There was no statistically significant difference in the density of neuropeptide Y+ neurons between subjects with autism and controls. The decreased calretinin+ density may disrupt the excitation/inhibition balance in the caudate leading to dysfunctional corticostriatal circuits. The description of such changes in autism spectrum disorder may clarify pathomechanisms and thereby help identify targets for drug intervention and novel therapeutic strategies.
Collapse
Affiliation(s)
- Istvan Adorjan
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.,Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Bashir Ahmed
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Virginia Feher
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Mario Torso
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Kristine Krug
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Margaret Esiri
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Steven A Chance
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Francis G Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
7
|
Hillis JM, Davies J, Mundim MV, Al-Dalahmah O, Szele FG. Cuprizone demyelination induces a unique inflammatory response in the subventricular zone. J Neuroinflammation 2016; 13:190. [PMID: 27550173 PMCID: PMC4994223 DOI: 10.1186/s12974-016-0651-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 07/04/2016] [Indexed: 12/04/2022] Open
Abstract
Background Cuprizone leads to demyelination of the corpus callosum (CC) and activates progenitor cells in the adjacent subventricular zone (SVZ), a stem cell niche which contributes to remyelination. The healthy SVZ contains semi-activated microglia and constitutively expresses the pro-inflammatory molecule galectin-3 (Gal-3) suggesting the niche uniquely regulates inflammation. Methods We studied the inflammatory response to cuprizone in the SVZ and CC in Gal-3 knockout mice using immunohistochemistry and with the in vitro neurosphere assay. Results Cuprizone caused loss of myelin basic protein (MBP) immunofluorescence in the CC suggesting demyelination. Cuprizone increased the density of CD45+/Iba1+ microglial cells and also increased Gal-3 expression in the CC. Surprisingly, the number of Gal-3+ and CD45+ cells decreased in the SVZ after cuprizone, suggesting inflammation was selectively reduced therein. Inflammation can regulate SVZ proliferation and indeed the number of phosphohistone H3+ (PHi3+) cells decreased in the SVZ but increased in the CC in both genotypes after cuprizone treatment. BrdU+ SVZ cell numbers also decreased in the SVZ after cuprizone, and this effect was significantly greater at 3 weeks in Gal-3−/− mice compared to WT, suggesting Gal-3 normally limits SVZ cell emigration following cuprizone treatment. Conclusions This study reveals a uniquely regulated inflammatory response in the SVZ and shows that Gal-3 participates in remyelination in the cuprizone model. This contrasts with more severe models of demyelination which induce SVZ inflammation and suggests the extent of demyelination affects the SVZ neurogenic response. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0651-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- James M Hillis
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
| | - Julie Davies
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
| | - Mayara Vieira Mundim
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK.,Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, 04039-032, Brazil
| | - Osama Al-Dalahmah
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
| | - Francis G Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK.
| |
Collapse
|
8
|
Höfling C, Kulesskaya N, Jaako K, Peltonen I, Männistö PT, Nurmi A, Vartiainen N, Morawski M, Zharkovsky A, Võikar V, Roßner S, García-Horsman JA. Deficiency of prolyl oligopeptidase in mice disturbs synaptic plasticity and reduces anxiety-like behaviour, body weight, and brain volume. Eur Neuropsychopharmacol 2016; 26:1048-61. [PMID: 26996375 DOI: 10.1016/j.euroneuro.2016.02.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 02/10/2016] [Accepted: 02/20/2016] [Indexed: 12/26/2022]
Abstract
Prolyl oligopeptidase (PREP) has been implicated in neurodegeneration and neuroinflammation and has been considered a drug target to enhance memory in dementia. However, the true physiological role of PREP is not yet understood. In this paper, we report the phenotyping of a mouse line where the PREP gene has been knocked out. This work indicates that the lack of PREP in mice causes reduced anxiety but also hyperactivity. The cortical volumes of PREP knockout mice were smaller than those of wild type littermates. Additionally, we found increased expression of diazepam binding inhibitor protein in the cortex and of the somatostatin receptor-2 in the hippocampus of PREP knockout mice. Furthermore, immunohistochemistry and tail suspension test revealed lack of response of PREP knockout mice to lipopolysaccharide insult. Further analysis revealed significantly increased levels of polysialylated-neural cell adhesion molecule in PREP deficient mice. These findings might be explained as possible alteration in brain plasticity caused by PREP deficiency, which in turn affect behaviour and brain development.
Collapse
Affiliation(s)
- Corinna Höfling
- Paul Flechsig Institute for Brain Research, University of Leipzig, Germany
| | | | - Külli Jaako
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia
| | - Iida Peltonen
- Division of Pharmacology and Pharmacotherapy, University of Helsinki, Finland
| | - Pekka T Männistö
- Division of Pharmacology and Pharmacotherapy, University of Helsinki, Finland
| | - Antti Nurmi
- Charles River Drug Discovery Services, Kuopio, Finland
| | | | - Markus Morawski
- Paul Flechsig Institute for Brain Research, University of Leipzig, Germany
| | - Alexander Zharkovsky
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia
| | | | - Steffen Roßner
- Paul Flechsig Institute for Brain Research, University of Leipzig, Germany
| | | |
Collapse
|
9
|
James RE, Hillis J, Adorján I, Gration B, Mundim MV, Iqbal AJ, Majumdar MM, Yates RL, Richards MMH, Goings GE, DeLuca GC, Greaves DR, Miller SD, Szele FG. Loss of galectin-3 decreases the number of immune cells in the subventricular zone and restores proliferation in a viral model of multiple sclerosis. Glia 2015; 64:105-21. [PMID: 26337870 DOI: 10.1002/glia.22906] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 08/06/2015] [Accepted: 08/07/2015] [Indexed: 12/30/2022]
Abstract
Multiple sclerosis (MS) frequently starts near the lateral ventricles, which are lined by subventricular zone (SVZ) progenitor cells that can migrate to lesions and contribute to repair. Because MS-induced inflammation may decrease SVZ proliferation and thus limit repair, we studied the role of galectin-3 (Gal-3), a proinflammatory protein. Gal-3 expression was increased in periventricular regions of human MS in post-mortem brain samples and was also upregulated in periventricular regions in a murine MS model, Theiler's murine encephalomyelitis virus (TMEV) infection. Whereas TMEV increased SVZ chemokine (CCL2, CCL5, CCL, and CXCL10) expression in wild type (WT) mice, this was inhibited in Gal-3(-/-) mice. Though numerous CD45+ immune cells entered the SVZ of WT mice after TMEV infection, their numbers were significantly diminished in Gal-3(-/-) mice. TMEV also reduced neuroblast and proliferative SVZ cell numbers in WT mice but this was restored in Gal-3(-/-) mice and was correlated with increased numbers of doublecortin+ neuroblasts in the corpus callosum. In summary, our data showed that loss of Gal-3 blocked chemokine increases after TMEV, reduced immune cell migration into the SVZ, reestablished SVZ proliferation and increased the number of progenitors in the corpus callosum. These results suggest Gal-3 plays a central role in modulating the SVZ neurogenic niche's response to this model of MS.
Collapse
Affiliation(s)
- Rachel E James
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, OX1 3HS, United Kingdom
| | - James Hillis
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, OX1 3HS, United Kingdom
| | - István Adorján
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, OX1 3HS, United Kingdom
| | - Betty Gration
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, OX1 3HS, United Kingdom
| | - Mayara V Mundim
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, OX1 3HS, United Kingdom
| | - Asif J Iqbal
- Dunn School of Pathology, University of Oxford, Oxford, OX1 3HS, United Kingdom
| | - Moon-Moon Majumdar
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, OX1 3HS, United Kingdom
| | - Richard L Yates
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, OX1 3HS, United Kingdom
| | - Maureen M H Richards
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Gwendolyn E Goings
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, OX1 3HS, United Kingdom
| | - David R Greaves
- Dunn School of Pathology, University of Oxford, Oxford, OX1 3HS, United Kingdom
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Francis G Szele
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, OX1 3HS, United Kingdom
| |
Collapse
|
10
|
Liu G, Rustom N, Litteljohn D, Bobyn J, Rudyk C, Anisman H, Hayley S. Use of induced pluripotent stem cell derived neurons engineered to express BDNF for modulation of stressor related pathology. Front Cell Neurosci 2014; 8:316. [PMID: 25352778 PMCID: PMC4196567 DOI: 10.3389/fncel.2014.00316] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 09/20/2014] [Indexed: 01/18/2023] Open
Abstract
Combined cell and gene-based therapeutic strategies offer potential in the treatment of neurodegenerative and psychiatric conditions that have been associated with structural brain disturbances. In the present investigation, we used a novel virus-free re-programming method to generate induced pluripotent stem cells (iPSCs), and then subsequently transformed these cells into neural cells which over-expressed brain derived neurotrophic factor (BDNF). Importantly, the infusion of iPSC derived neural cells (as a cell replacement and gene delivery tool) and BDNF (as a protective factor) influenced neuronal outcomes. Specifically, intracerebroventricular transplantation of iPSC-derived neural progenitors that over-expressed BDNF reversed the impact of immune (lipopolysaccharide) and chronic stressor challenges upon subventricular zone adult neurogenesis, and the iPSC-derived neural progenitor cells alone blunted the stressor-induced corticosterone response. Moreover, our findings indicate that mature dopamine producing neurons can be generated using iPSC procedures and appear to be viable when infused in vivo. Taken together, these data could have important implications for using gene-plus-cell replacement methods to modulate stressor related pathology.
Collapse
Affiliation(s)
- Gele Liu
- Hayley Lab, Department of Neuroscience, Carleton University Ottawa, ON, Canada
| | - Nazneen Rustom
- Hayley Lab, Department of Neuroscience, Carleton University Ottawa, ON, Canada
| | - Darcy Litteljohn
- Hayley Lab, Department of Neuroscience, Carleton University Ottawa, ON, Canada
| | - Jessica Bobyn
- Hayley Lab, Department of Neuroscience, Carleton University Ottawa, ON, Canada
| | - Chris Rudyk
- Hayley Lab, Department of Neuroscience, Carleton University Ottawa, ON, Canada
| | - Hymie Anisman
- Hayley Lab, Department of Neuroscience, Carleton University Ottawa, ON, Canada
| | - Shawn Hayley
- Hayley Lab, Department of Neuroscience, Carleton University Ottawa, ON, Canada
| |
Collapse
|
11
|
Young CC, Al-Dalahmah O, Lewis NJ, Brooks KJ, Jenkins MM, Poirier F, Buchan AM, Szele FG. Blocked angiogenesis in Galectin-3 null mice does not alter cellular and behavioral recovery after middle cerebral artery occlusion stroke. Neurobiol Dis 2013; 63:155-64. [PMID: 24269916 DOI: 10.1016/j.nbd.2013.11.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 10/24/2013] [Accepted: 11/12/2013] [Indexed: 12/31/2022] Open
Abstract
Angiogenesis is thought to decrease stroke size and improve behavioral outcomes and therefore several clinical trials are seeking to augment it. Galectin-3 (Gal-3) expression increases after middle cerebral artery occlusion (MCAO) and has been proposed to limit damage 3days after stroke. We carried out mild MCAO that damages the striatum but spares the cerebral cortex and SVZ. Gal-3 gene deletion prevented vascular endothelial growth factor (VEGF) upregulation after MCAO. This inhibited post-MCAO increases in endothelial proliferation and angiogenesis in the striatum allowing us to uniquely address the function of angiogenesis in this model of stroke. Apoptosis and infarct size were unchanged in Gal-3(-/-) mice 7 and 14 days after MCAO, suggesting that angiogenesis does not affect lesion size. Microglial and astrocyte activation/proliferation after MCAO was similar in wild type and Gal-3(-/-) mice. In addition, openfield activity, motor hemiparesis, proprioception, reflex, tremors and grooming behaviors were essentially identical between WT and Gal-3(-/-) mice at 1, 3, 7, 10 and 14 days after MCAO, suggesting that penumbral angiogenesis has limited impact on behavioral recovery. In addition to angiogenesis, increased adult subventricular zone (SVZ) neurogenesis is thought to provide neuroprotection after stroke in animal models. SVZ neurogenesis and migration to lesion were overall unaffected by the loss of Gal-3, suggesting no compensation for the lack of angiogenesis in Gal-3(-/-) mice. Because angiogenesis and neurogenesis are usually coordinately regulated, identifying their individual effects on stroke has hitherto been difficult. These results show that Gal-3 is necessary for angiogenesis in stroke in a VEGF-dependant manner, but suggest that angiogenesis may be dispensable for post-stroke endogenous repair, therefore drawing into question the clinical utility of augmenting angiogenesis.
Collapse
Affiliation(s)
- Christopher C Young
- University of Oxford, Department of Physiology, Anatomy and Genetics, University of Oxford, OX1 3QX, UK
| | - Osama Al-Dalahmah
- University of Oxford, Department of Physiology, Anatomy and Genetics, University of Oxford, OX1 3QX, UK
| | - Nicola J Lewis
- University of Oxford, Department of Physiology, Anatomy and Genetics, University of Oxford, OX1 3QX, UK
| | - Keith J Brooks
- Nuffield Department of Clinical Medicine, University of Oxford, OX1 3QX, UK
| | - Micaela M Jenkins
- Nuffield Department of Clinical Medicine, University of Oxford, OX1 3QX, UK
| | - Françoise Poirier
- Institut Jacques Monod, UMR CNRS 7592, Université Paris Diderot, 75205 Paris 13, France
| | - Alastair M Buchan
- Nuffield Department of Clinical Medicine, University of Oxford, OX1 3QX, UK
| | - Francis G Szele
- University of Oxford, Department of Physiology, Anatomy and Genetics, University of Oxford, OX1 3QX, UK.
| |
Collapse
|
12
|
Mobilization of progenitors in the subventricular zone to undergo oligodendrogenesis in the Theiler's virus model of multiple sclerosis: implications for remyelination at lesions sites. Exp Neurol 2013; 250:348-52. [PMID: 24148569 DOI: 10.1016/j.expneurol.2013.10.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 10/08/2013] [Accepted: 10/14/2013] [Indexed: 12/21/2022]
Abstract
Remyelination involves the generation of new myelin sheaths around axons, as occurs spontaneously in many multiple sclerosis (MS) lesions and other demyelinating diseases. When considering repairing a diseased brain, the adult mouse subventricular zone (SVZ) is of particular interest since the stem cells in this area can migrate and differentiate into the three major cell types in the central nervous system (CNS). In Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD), we assessed the relative contribution of the SVZ to the remyelination in the corpus callosum at preclinical stages in this MS model. CNPase, MBP and Luxol Fast Blue staining revealed prominent demyelination 35days post-infection (dpi), concomitant with a strong staining in GFAP(+) type B astrocytes in the SVZ and the increased proliferation in this area. The migration of oligodendrocyte progenitors from the SVZ contributed to the remyelination observed at 60 dpi, evident through the number of APC(+)/BrdU(+) mature oligodendrocytes in the corpus callosum of infected animals. These data suggest that the inflammation induced by the Theiler's virus not only provokes strong preclinical demyelination but also, it is correlated with oligodendrocyte generation in the adult SVZ, cells that along with resident progenitor cells contribute to the prompt remyelination observed in the corpus callosum.
Collapse
|
13
|
Jafari M, Haist V, Baumgärtner W, Wagner S, Stein VM, Tipold A, Wendt H, Potschka H. Impact of Theiler's virus infection on hippocampal neuronal progenitor cells: differential effects in two mouse strains. Neuropathol Appl Neurobiol 2013; 38:647-64. [PMID: 22288387 DOI: 10.1111/j.1365-2990.2012.01256.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIMS Disease-associated alterations in hippocampal neurogenesis are discussed as an important factor contributing to long-term consequences of central nervous system diseases. Therefore, the study aimed to determine the impact of Theiler's murine encephalomyelitis virus infection on hippocampal cell proliferation, neuronal progenitor cells and neurogenesis as well as the influence of microglia on respective disease-associated alterations. METHODS The impact of the infection was evaluated in two mouse strains which differ in the disease course, with an acute polioencephalitis followed by virus elimination in C57BL/6 mice and a chronic demyelinating disease in SJL/J mice. RESULTS Infection with the low neurovirulent BeAn strain did not exert significant acute effects regardless of the mouse strain. In the chronic phase, the number of neuronal progenitor cells and early postmitotic neurones was significantly reduced in infected SJL/J mice, whereas no long-term alterations were observed in C57BL/6 mice. A contrasting course of microglia activation was observed in the two mouse strains, with an early increase in the number of activated microglia cells in SJL/J mice and a delayed increase in C57BL/6 mice. Quantitative analysis did not confirm a correlation between the number of activated microglia and the number of neuronal progenitor cells and early postmitotic neurones. However, flow cytometric analyses revealed alterations in the functional state of microglial cells which might have affected the generation of neuronal progenitor cells. CONCLUSIONS Theiler's murine encephalomyelitis virus infection can exert delayed effects on the hippocampal neuronal progenitor population with long-term alterations evident 3 months following infection. These alterations proved to depend on strain susceptibility and might contribute to detrimental consequences of virus encephalitis such as cognitive impairment.
Collapse
Affiliation(s)
- M Jafari
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Mecha M, Carrillo-Salinas FJ, Mestre L, Feliú A, Guaza C. Viral models of multiple sclerosis: neurodegeneration and demyelination in mice infected with Theiler's virus. Prog Neurobiol 2013; 101-102:46-64. [PMID: 23201558 PMCID: PMC7117056 DOI: 10.1016/j.pneurobio.2012.11.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 10/30/2012] [Accepted: 11/12/2012] [Indexed: 11/02/2022]
Abstract
Multiple sclerosis (MS) is a complex inflammatory disease of unknown etiology that affects the central nervous system (CNS) white matter, and for which no effective cure exists. Indeed, whether the primary event in MS pathology affects myelin or axons of the CNS remains unclear. Animal models are necessary to identify the immunopathological mechanisms involved in MS and to develop novel therapeutic and reparative approaches. Specifically, viral models of chronic demyelination and axonal damage have been used to study the contribution of viruses in human MS, and they have led to important breakthroughs in our understanding of MS pathology. The Theiler's murine encephalomyelitis virus (TMEV) model is one of the most commonly used MS models, although other viral models are also used, including neurotropic strains of mouse hepatitis virus (MHV) that induce chronic inflammatory demyelination with similar histological features to those observed in MS. This review will discuss the immunopathological mechanisms involved in TMEV-induced demyelinating disease (TMEV-IDD). The TMEV model reproduces a chronic progressive disease due to the persistence of the virus for the entire lifespan in susceptible mice. The evolution and significance of the axonal damage and neuroinflammation, the importance of epitope spread from viral to myelin epitopes, the presence of abortive remyelination and the existence of a brain pathology in addition to the classical spinal cord demyelination, are some of the findings that will be discussed in the context of this TMEV-IDD model. Despite their limitations, viral models remain an important tool to study the etiology of MS, and to understand the clinical and pathological variability associated with this disease.
Collapse
Key Words
- ab, antibody
- ag, antigen
- apc, antigen presenting cell
- bbb, blood–brain barrier
- cns, central nervous system
- cox-2, cyclooxygenase-2
- ctl, cytotoxic t lymphocytes
- dpi, days post-infection
- da, daniels strain of theiler's virus
- eae, experimental autoimmune encephalomyelitis
- galc, galactocerebroside
- mbp, myelin basic protein
- mnc, mononuclear cells
- mhc, major histocompatibility complex
- mhv, mouse hepatitis virus
- mog, myelin oligodendrocyte glycoprotein
- ms, multiple sclerosis
- naa, n-acetylaspartate
- no, nitric oxide
- pcr, polymerase chain reaction
- plp, myelin proteolipid protein
- pprs, pattern recognition receptors
- sfv, semliki forest virus
- sv, sindbis virus
- tmev, theiler's murine encephalomyelitis virus
- tmev-idd, theiler's murine encephalomyelitis virus-induced demyelinating disease
- tregs, regulatory t cells
- theiler's virus
- multiple sclerosis
- demyelination
- axonal damage
- neuroinflammation
- spinal cord pathology
- brain pathology
Collapse
Affiliation(s)
| | | | | | | | - Carmen Guaza
- Neuroimmunology Group, Functional and System Neurobiology Department, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Avda Dr Arce 37, 28002 Madrid, Spain
| |
Collapse
|
15
|
Kummerfeld M, Seehusen F, Klein S, Ulrich R, Kreutzer R, Gerhauser I, Herder V, Baumgärtner W, Beineke A. Periventricular demyelination and axonal pathology is associated with subependymal virus spread in a murine model for multiple sclerosis. Intervirology 2012; 55:401-16. [PMID: 22538300 DOI: 10.1159/000336563] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 01/02/2012] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Theiler's murine encephalomyelitis virus (TMEV) infection of mice is a widely used animal model for demyelinating disorders, such as multiple sclerosis (MS). The aim of the present study was to identify topographical differences of TMEV spread and demyelination in the brain of experimentally infected susceptible SJL/J mice and resistant C57BL/6 mice. METHODS Demyelination was confirmed by Luxol fast blue and cresyl violet staining and axonal damage by neurofilament-specific and β-amyloid precursor protein-specific immunohistochemistry. Viral dissemination within the central nervous system (CNS) was quantified by immunohistochemistry and in situ hybridization. Further, the phenotype of infected cells was determined by confocal laser scanning microscopy. RESULTS An early transient infection of periventricular cells followed by demyelination and axonopathies around the fourth ventricle in SJL/J mice was noticed. Periventricular and brain stem demyelination was associated with a predominant infection of microglia/macrophages and oligodendrocytes. CONCLUSIONS Summarized, the demonstration of ependymal infection and subjacent spread into the brain parenchyma as well as regional virus clearance despite ongoing demyelination and axonal damage in other CNS compartments allows new insights into TME pathogenesis. This novel aspect of TMEV CNS interaction will enhance the understanding of region-specific susceptibilities to injury and regenerative capacities of the brain in this MS model.
Collapse
Affiliation(s)
- Maren Kummerfeld
- Department of Pathology, University of Veterinary Medicine Hanover, Hanover, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Young CC, Brooks KJ, Buchan AM, Szele FG. Cellular and molecular determinants of stroke-induced changes in subventricular zone cell migration. Antioxid Redox Signal 2011; 14:1877-88. [PMID: 20673127 PMCID: PMC3078507 DOI: 10.1089/ars.2010.3435] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A remarkable aspect of adult neurogenesis is that the tight regulation of subventricular zone (SVZ) neuroblast migration is altered after ischemic stroke and newborn neurons emigrate towards the injury. This phenomenon is an essential component of endogenous repair and also serves to illuminate normal mechanisms and rules that govern SVZ migration. Stroke causes inflammation that leads to cytokine and chemokine release, and SVZ neuroblasts that express their receptors are recruited. Metalloproteinases create pathways and new blood vessels provide a scaffold to facilitate neuroblast migration between the SVZ and the infarct. Most experiments have studied the peri-lesion parenchyma and relatively little is known about SVZ remodeling after stroke. Migration in the SVZ is tightly regulated by cellular interactions and molecular signaling; how are these altered after stroke to allow emigration? Do ependymal cells contribute to this process, given their reported neurogenic potential? How does stroke affect ependymal cell regulation of cerebrospinal fluid flow? Given the heterogeneity of SVZ progenitors, do all types of neuroblasts migrate out, or is this confined to specific subtypes of cells? We discuss these and other questions in our review and propose experiments to address them.
Collapse
Affiliation(s)
- Christopher C Young
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | | | | | | |
Collapse
|
17
|
Silvestroff L, Bartucci S, Soto E, Gallo V, Pasquini J, Franco P. Cuprizone-induced demyelination in CNP::GFP transgenic mice. J Comp Neurol 2010; 518:2261-83. [PMID: 20437527 DOI: 10.1002/cne.22330] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Cuprizone (bis-cyclohexanone oxaldihydrazone) was previously shown to induce demyelination in white matter enriched brain structures. In the present study we used the cuprizone demyelination model in transgenic mice expressing the enhanced green fluorescent protein (GFP) under the 2'-3'-cyclic nucleotide 3'-phosphodiesterase (CNPase) promoter. The use of these particular transgenic mice allows easy detection of cells belonging to the entire oligodendroglial (OLG) lineage, ranging from OLG precursors to mature myelinating OLGs. We were able to evaluate the precise extent of oligodendroglial cell damage and recovery within the murine adult central nervous system (CNS) after inducing demyelination by acute cuprizone intoxication. A generalized loss of GFP+ cells was observed after cuprizone exposure and correlated with a decline in myelin basic protein (MBP) expression. OLGs were depleted in many brain areas that were previously thought to be unaffected by cuprizone treatment. Thus, in addition to the well-known cuprizone effects on the medial corpus callosum, we also found a loss of GFP+ cells in most brain structures, particularly in the caudatus putamen, cortex, anterior commissure, olfactory bulb, hippocampus, optic chiasm, brainstem, and cingulum. Loss of GFP+ cells was accompanied by extensive astrogliosis and microglial activation, although neurons were not affected. Interestingly, cuprizone-treated animals showed both activation of GFAP expression and a higher proliferation rate in subventricular zone cells. A week after cuprizone removal from the diet, GFP+ oligodendroglial cells began repopulating the damaged structures. GFP expression precedes that of MBP and allows OLG detection before myelin restoration.
Collapse
Affiliation(s)
- Lucas Silvestroff
- Departamento de Química Biológica, IQUIFIB and IIMHNO, Facultad de Farmacia y Bioquímica, UBA-CONICET, Junín 956, Buenos Aires C1113AAD, Argentina
| | | | | | | | | | | |
Collapse
|