1
|
Dedoni S, Scherma M, Camoglio C, Siddi C, Dazzi L, Puliga R, Frau J, Cocco E, Fadda P. An overall view of the most common experimental models for multiple sclerosis. Neurobiol Dis 2023:106230. [PMID: 37453561 DOI: 10.1016/j.nbd.2023.106230] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 07/01/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023] Open
Abstract
Multiple sclerosis (MS) is a complex chronic disease with an unknown etiology. It is considered an inflammatory demyelinating and neurodegenerative disorder of the central nervous system (CNS) characterized, in most cases, by an unpredictable onset of relapse and remission phases. The disease generally starts in subjects under 40; it has a higher incidence in women and is described as a multifactorial disorder due to the interaction between genetic and environmental risk factors. Unfortunately, there is currently no definitive cure for MS. Still, therapies can modify the disease's natural history, reducing the relapse rate and slowing the progression of the disease or managing symptoms. The limited access to human CNS tissue slows down. It limits the progression of research on MS. This limit has been partially overcome over the years by developing various experimental models to study this disease. Animal models of autoimmune demyelination, such as experimental autoimmune encephalomyelitis (EAE) and viral and toxin or transgenic MS models, represent the most significant part of MS research approaches. These models have now been complemented by ex vivo studies, using organotypic brain slice cultures and in vitro, through induced Pluripotent Stem cells (iPSCs). We will discuss which clinical features of the disorders might be reproduced and investigated in vivo, ex vivo, and in vitro in models commonly used in MS research to understand the processes behind the neuropathological events occurring in the CNS of MS patients. The primary purpose of this review is to give the reader a global view of the main paradigms used in MS research, spacing from the classical animal models to transgenic mice and 2D and 3D cultures.
Collapse
Affiliation(s)
- S Dedoni
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - M Scherma
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - C Camoglio
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - C Siddi
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy
| | - L Dazzi
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato (Cagliari), Italy.
| | - R Puliga
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato (Cagliari), Italy.
| | - J Frau
- Regional Multiple Sclerosis Center, ASSL Cagliari, ATS Sardegna, Italy
| | - E Cocco
- Regional Multiple Sclerosis Center, ASSL Cagliari, ATS Sardegna, Italy; Department Medical Science and Public Health, University of Cagliari, Italy.
| | - P Fadda
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy; Neuroscience Institute, Section of Cagliari, National Research Council of Italy (CNR), Cagliari, Italy.
| |
Collapse
|
2
|
Perez-Gianmarco L, Kurt B, Kukley M. Technical approaches and challenges to study AMPA receptors in oligodendrocyte lineage cells: Past, present, and future. Glia 2023; 71:819-847. [PMID: 36453615 DOI: 10.1002/glia.24305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 11/05/2022] [Accepted: 11/10/2022] [Indexed: 12/03/2022]
Abstract
Receptors for α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPARs) are ligand-gated ionotropic receptors for glutamate that is a major excitatory neurotransmitter in the central nervous system. AMPARs are located at postsynaptic sites of neuronal synapses where they mediate fast synaptic signaling and synaptic plasticity. Remarkably, AMPARs are also expressed by glial cells. Their expression by the oligodendrocyte (OL) lineage cells is of special interest because AMPARs mediate fast synaptic communication between neurons and oligodendrocyte progenitor cells (OPCs), modulate proliferation and differentiation of OPCs, and may also be involved in regulation of myelination. On the other hand, during pathological conditions, AMPARs may mediate damage of the OL lineage cells. In the present review, we focus on the technical approaches that have been used to study AMPARs in the OL lineage cells, and discuss future perspectives of AMPAR research in these glial cells.
Collapse
Affiliation(s)
- Lucila Perez-Gianmarco
- Laboratory of Neuronal and Glial Physiology, Achucarro Basque Center for Neuroscience, Leioa, Spain.,Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Begüm Kurt
- Laboratory of Neuronal and Glial Physiology, Achucarro Basque Center for Neuroscience, Leioa, Spain.,Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Maria Kukley
- Laboratory of Neuronal and Glial Physiology, Achucarro Basque Center for Neuroscience, Leioa, Spain.,Ikerbasque - Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
3
|
Bayón-Cordero L, Ochoa-Bueno BI, Ruiz A, Ozalla M, Matute C, Sánchez-Gómez MV. GABA Receptor Agonists Protect From Excitotoxic Damage Induced by AMPA in Oligodendrocytes. Front Pharmacol 2022; 13:897056. [PMID: 35959434 PMCID: PMC9360600 DOI: 10.3389/fphar.2022.897056] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/06/2022] [Indexed: 11/17/2022] Open
Abstract
Oligodendrocytes are the myelin forming cells of the central nervous system, and their vulnerability to excitotoxicity induced by glutamate contributes to the pathogenesis of neurological disorders including brain ischemia and neurodegenerative diseases, such as multiple sclerosis. In addition to glutamate receptors, oligodendrocytes express GABA receptors (GABAR) that are involved in their survival and differentiation. The interactions between glutamate and GABAergic systems are well documented in neurons, under both physiological and pathological conditions, but this potential crosstalk in oligodendrocytes has not been studied in depth. Here, we evaluated the protective effect of GABAR agonists, baclofen (GABAB) and muscimol (GABAA), against AMPA-induced excitotoxicity in cultured rat oligodendrocytes. First, we observed that both baclofen and muscimol reduced cell death and caspase-3 activation after AMPA insult, proving their oligoprotective potential. Interestingly, analysis of the cell-surface expression of calcium-impermeable GluR2 subunits in oligodendrocytes revealed that GABAergic agonists significantly reverted GluR2 internalization induced by AMPA. We determined that baclofen and muscimol also impaired AMPA-induced intracellular calcium increase and subsequent mitochondrial membrane potential alteration, ROS generation, and calpain activation. However, AMPA-triggered activation of Src, Akt, JNK and CREB was not affected by baclofen or muscimol. Overall, our results suggest that GABAR activation initiates alternative molecular mechanisms that attenuate AMPA-mediated apoptotic excitotoxicity in oligodendrocytes by interfering with expression of GluR subunits in membranes and with calcium-dependent intracellular signaling pathways. Together, these findings provide evidence of GABAR agonists as potential oligodendroglial protectants in central nervous system disorders.
Collapse
Affiliation(s)
- Laura Bayón-Cordero
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Blanca Isabel Ochoa-Bueno
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Asier Ruiz
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Marina Ozalla
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Carlos Matute
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - María Victoria Sánchez-Gómez
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| |
Collapse
|
4
|
Ransom BR, Goldberg MP, Arai K, Baltan S. White Matter Pathophysiology. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00009-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
5
|
Fairless R, Bading H, Diem R. Pathophysiological Ionotropic Glutamate Signalling in Neuroinflammatory Disease as a Therapeutic Target. Front Neurosci 2021; 15:741280. [PMID: 34744612 PMCID: PMC8567076 DOI: 10.3389/fnins.2021.741280] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/30/2021] [Indexed: 01/15/2023] Open
Abstract
Glutamate signalling is an essential aspect of neuronal communication involving many different glutamate receptors, and underlies the processes of memory, learning and synaptic plasticity. Despite neuroinflammatory diseases covering a range of maladies with very different biological causes and pathophysiologies, a central role for dysfunctional glutamate signalling is becoming apparent. This is not just restricted to the well-described role of glutamate in mediating neurodegeneration, but also includes a myriad of other influences that glutamate can exert on the vasculature, as well as immune cell and glial regulation, reflecting the ability of neurons to communicate with these compartments in order to couple their activity with neuronal requirements. Here, we discuss the role of pathophysiological glutamate signalling in neuroinflammatory disease, using both multiple sclerosis and Alzheimer's disease as examples, and how current steps are being made to harness our growing understanding of these processes in the development of neuroprotective strategies. This review focuses in particular on N-methyl-D-aspartate (NMDA) and 2-amino-3-(3-hydroxy-5-methylisooxazol-4-yl) propionate (AMPA) type ionotropic glutamate receptors, although metabotropic, G-protein-coupled glutamate receptors may also contribute to neuroinflammatory processes. Given the indispensable roles of glutamate-gated ion channels in synaptic communication, means of pharmacologically distinguishing between physiological and pathophysiological actions of glutamate will be discussed that allow deleterious signalling to be inhibited whilst minimising the disturbance of essential neuronal function.
Collapse
Affiliation(s)
- Richard Fairless
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Ricarda Diem
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
6
|
Roth LM, Akay-Espinoza C, Grinspan JB, Jordan-Sciutto KL. HIV-induced neuroinflammation inhibits oligodendrocyte maturation via glutamate-dependent activation of the PERK arm of the integrated stress response. Glia 2021; 69:2252-2271. [PMID: 34058792 DOI: 10.1002/glia.24033] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 12/15/2022]
Abstract
Despite combined antiretroviral therapy (cART), HIV-associated neurocognitive disorder (HAND) affects 30-50% of HIV-positive patients. Importantly, persistent white matter pathologies, specifically corpus callosum thinning and disruption of white matter microstructures observed in patients with HAND despite viral control through cART, raise the possibility that HIV infection in the setting of suboptimal cART may perturb oligodendrocyte (OL) maturation, function and/or survival, influencing HAND persistence in the cART era. To examine the effect of HIV infection on OL maturation, we used supernatants of primary human monocyte-derived macrophages infected with HIV (HIV/MDMs) to treat primary cultures of rat oligodendrocyte precursor cells (OPCs) during their differentiation to mature OLs. Using immunostaining for lineage-specific markers, we found that HIV/MDMs significantly inhibited OPC maturation. Based on our previous studies, we examined the potential role of several signaling pathways, including ionotropic glutamate receptors and the integrated stress response (ISR), and found that AMPA receptors (AMPAR)/kainic acid (KA) receptors (KARs) mediated the HIV/MDMs-induced defect in OL maturation. We also found that the treatment of OPC cultures with glutamate or AMPAR/KAR agonists phenocopied this effect. Blocking ISR activation, specifically the PERK arm of the ISR, protected OPCs from HIV/MDMs-mediated inhibition of OL maturation. Further, while glutamate, AMPA, and KA activated the ISR, inhibition of AMPAR/KAR activation prevented ISR induction in OPCs and rescued OL maturation. Collectively, these data identify glutamate signaling via ISR activation as a potential therapeutic pathway to ameliorate white matter pathologies in HAND and highlight the need for further investigation of their contribution to cognitive impairment.
Collapse
Affiliation(s)
- Lindsay M Roth
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Neurology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Cagla Akay-Espinoza
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Judith B Grinspan
- Department of Neurology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Kelly L Jordan-Sciutto
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
Choi DW. Excitotoxicity: Still Hammering the Ischemic Brain in 2020. Front Neurosci 2020; 14:579953. [PMID: 33192266 PMCID: PMC7649323 DOI: 10.3389/fnins.2020.579953] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
Interest in excitotoxicity expanded following its implication in the pathogenesis of ischemic brain injury in the 1980s, but waned subsequent to the failure of N-methyl-D-aspartate (NMDA) antagonists in high profile clinical stroke trials. Nonetheless there has been steady progress in elucidating underlying mechanisms. This review will outline the historical path to current understandings of excitotoxicity in the ischemic brain, and suggest that this knowledge should be leveraged now to develop neuroprotective treatments for stroke.
Collapse
Affiliation(s)
- Dennis W Choi
- Department of Neurology, SUNY Stony Brook, Stony Brook, NY, United States
| |
Collapse
|
8
|
Zimmer TS, Broekaart DWM, Gruber VE, van Vliet EA, Mühlebner A, Aronica E. Tuberous Sclerosis Complex as Disease Model for Investigating mTOR-Related Gliopathy During Epileptogenesis. Front Neurol 2020; 11:1028. [PMID: 33041976 PMCID: PMC7527496 DOI: 10.3389/fneur.2020.01028] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022] Open
Abstract
Tuberous sclerosis complex (TSC) represents the prototypic monogenic disorder of the mammalian target of rapamycin (mTOR) pathway dysregulation. It provides the rational mechanistic basis of a direct link between gene mutation and brain pathology (structural and functional abnormalities) associated with a complex clinical phenotype including epilepsy, autism, and intellectual disability. So far, research conducted in TSC has been largely neuron-oriented. However, the neuropathological hallmarks of TSC and other malformations of cortical development also include major morphological and functional changes in glial cells involving astrocytes, oligodendrocytes, NG2 glia, and microglia. These cells and their interglial crosstalk may offer new insights into the common neurobiological mechanisms underlying epilepsy and the complex cognitive and behavioral comorbidities that are characteristic of the spectrum of mTOR-associated neurodevelopmental disorders. This review will focus on the role of glial dysfunction, the interaction between glia related to mTOR hyperactivity, and its contribution to epileptogenesis in TSC. Moreover, we will discuss how understanding glial abnormalities in TSC might give valuable insight into the pathophysiological mechanisms that could help to develop novel therapeutic approaches for TSC or other pathologies characterized by glial dysfunction and acquired mTOR hyperactivation.
Collapse
Affiliation(s)
- Till S Zimmer
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Diede W M Broekaart
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | | | - Erwin A van Vliet
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| | - Angelika Mühlebner
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, Netherlands
| |
Collapse
|
9
|
Kula B, Chen T, Kukley M. Glutamatergic signaling between neurons and oligodendrocyte lineage cells: Is it synaptic or non‐synaptic? Glia 2019; 67:2071-2091. [DOI: 10.1002/glia.23617] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/12/2019] [Accepted: 03/18/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Bartosz Kula
- Group of Neuron Glia InteractionUniversity of Tübingen Tübingen Germany
- Graduate Training Centre for NeuroscienceUniversity of Tübingen Tübingen Germany
| | - Ting‐Jiun Chen
- Center for Neuroscience ResearchChildren's Research Institute, Children's National Medical Center Washington District of Columbia
| | - Maria Kukley
- Group of Neuron Glia InteractionUniversity of Tübingen Tübingen Germany
- Research Institute for OphthalmologyUniversity Hospital Tübingen Tübingen Germany
| |
Collapse
|
10
|
Hu M, Hong L, Liu C, Hong S, He S, Zhou M, Huang G, Chen Q. Electrical stimulation enhances neuronal cell activity mediated by Schwann cell derived exosomes. Sci Rep 2019; 9:4206. [PMID: 30862846 PMCID: PMC6414536 DOI: 10.1038/s41598-019-41007-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 02/27/2019] [Indexed: 12/13/2022] Open
Abstract
Electrical stimulation (ES) therapy has good effects in patients with nervous system injury-related diseases. ES promotes nerve cell regeneration and stimulates Schwann cells to express neurotrophic factors. The incidence of stress urinary incontinence (SUI) among elderly people is increasing. Some studies suggest that damage to the pudendal nerve is closely related to the pathogenesis of SUI. It has also been found that pelvic ES can reduce SUI symptoms in a rat model of SUI caused by pudendal nerve injury. Clinically, pelvic floor electrical stimulation is effective in patients with mild to moderate SUI. These studies indicate that ES may ameliorate damage to the pudendal nerve and thus achieve the goal of SUI treatment, although the mechanism of action of this treatment remains unclear. Therefore, the purpose of the present study was to clarify the relationships among ES, neural cells and Schwann cells at the cellular level. We applied ES to nerve cells at 100 mV/mm or 200 mV/mm for 0, 0.5, 1, or 2 h to investigate changes in nerve cell activity. We then co-cultured the nerve cells with Schwann cells to explore the influence of single-culture and co-culture conditions on the nerve cells. Compared to non-ES, ES of the nerve cells increased their activity. Compared to those in single culture, co-cultured nerve cells exhibited an additional increase in activity. We also found that Schwann cell derived exosomes could promote the activity of nerve cells, with glutamate and calcium ions playing a potential role in this process. These results suggest that the mutual regulation of neural cells and Schwann cells plays an important role in the process by which ES ameliorates neurological function, which may provide a basis for subsequent studies.
Collapse
Affiliation(s)
- Ming Hu
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, China
| | - Li Hong
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, China.
| | - Cheng Liu
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, China
| | - Shasha Hong
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, China
| | - Songming He
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, China
| | - Min Zhou
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, China
| | - Guotao Huang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, China
| | - Qian Chen
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, China
| |
Collapse
|
11
|
Canedo-Antelo M, Serrano MP, Manterola A, Ruiz A, Llavero F, Mato S, Zugaza JL, Pérez-Cerdá F, Matute C, Sánchez-Gómez MV. Inhibition of Casein Kinase 2 Protects Oligodendrocytes From Excitotoxicity by Attenuating JNK/p53 Signaling Cascade. Front Mol Neurosci 2018; 11:333. [PMID: 30271323 PMCID: PMC6146035 DOI: 10.3389/fnmol.2018.00333] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/27/2018] [Indexed: 12/12/2022] Open
Abstract
Oligodendrocytes are highly vulnerable to glutamate excitotoxicity, a central mechanism involved in tissue damage in Multiple Sclerosis (MS). Sustained activation of AMPA receptors in rat oligodendrocytes induces cytosolic calcium overload, mitochondrial depolarization, increase of reactive oxygen species, and activation of intracelular pathways resulting in apoptotic cell death. Although many signals driven by excitotoxicity have been identified, some of the key players are still under investigation. Casein kinase 2 (CK2) is a serine/threonine kinase, constitutively expressed in all eukaryotic tissues, involved in cell proliferation, malignant transformation and apoptosis. In this study, we identify CK2 as a critical regulator of oligodendrocytic death pathways and elucidate its role as a signal inductor following excitotoxic insults. We provide evidence that CK2 activity is up-regulated in AMPA-treated oligodendrocytes and CK2 inhibition significantly diminished AMPA receptor-induced oligodendroglial death. In addition, we analyzed mitogen-activated protein kinase (MAPK) signaling after excitotoxic insult. We observed that AMPA receptor activation induced a rapid increase in c-Jun N-terminal kinase (JNK) and p38 phosphorylation that was reduced after CK2 inhibition. Moreover, blocking their phosphorylation, we enhanced oligodendrocyte survival after excitotoxic insult. Finally, we observed that the tumor suppressor p53 is activated during AMPA receptor-induced cell death and, interestingly, down-regulated by JNK or CK2 inhibition. Together, these data indicate that the increase in CK2 activity induced by excitotoxic insults regulates MAPKs, triggers p53 activation and mediates subsequent oligodendroglial loss. Therefore, targeting CK2 may be a useful strategy to prevent oligodendrocyte death in MS and other diseases involving central nervous system (CNS) white matter.
Collapse
Affiliation(s)
- Manuel Canedo-Antelo
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain.,Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Mari Paz Serrano
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain.,Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Andrea Manterola
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain.,Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Asier Ruiz
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain.,Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Francisco Llavero
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Genética, Antropología Física y Fisiología Animal, Universidad del País Vasco (UPV/EHU), Leioa, Spain
| | - Susana Mato
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain.,Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - José Luis Zugaza
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Genética, Antropología Física y Fisiología Animal, Universidad del País Vasco (UPV/EHU), Leioa, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Fernando Pérez-Cerdá
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain.,Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Carlos Matute
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain.,Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - María Victoria Sánchez-Gómez
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain.,Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| |
Collapse
|
12
|
Barger SW. Gene regulation and genetics in neurochemistry, past to future. J Neurochem 2016; 139 Suppl 2:24-57. [PMID: 27747882 DOI: 10.1111/jnc.13629] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 03/01/2016] [Accepted: 03/30/2016] [Indexed: 12/14/2022]
Abstract
Ask any neuroscientist to name the most profound discoveries in the field in the past 60 years, and at or near the top of the list will be a phenomenon or technique related to genes and their expression. Indeed, our understanding of genetics and gene regulation has ushered in whole new systems of knowledge and new empirical approaches, many of which could not have even been imagined prior to the molecular biology boon of recent decades. Neurochemistry, in the classic sense, intersects with these concepts in the manifestation of neuropeptides, obviously dependent upon the central dogma (the established rules by which DNA sequence is eventually converted into protein primary structure) not only for their conformation but also for their levels and locales of expression. But, expanding these considerations to non-peptide neurotransmitters illustrates how gene regulatory events impact neurochemistry in a much broader sense, extending beyond the neurochemicals that translate electrical signals into chemical ones in the synapse, to also include every aspect of neural development, structure, function, and pathology. From the beginning, the mutability - yet relative stability - of genes and their expression patterns were recognized as potential substrates for some of the most intriguing phenomena in neurobiology - those instances of plasticity required for learning and memory. Near-heretical speculation was offered in the idea that perhaps the very sequence of the genome was altered to encode memories. A fascinating component of the intervening progress includes evidence that the central dogma is not nearly as rigid and consistent as we once thought. And this mutability extends to the potential to manipulate that code for both experimental and clinical purposes. Astonishing progress has been made in the molecular biology of neurochemistry during the 60 years since this journal debuted. Many of the gains in conceptual understanding have been driven by methodological progress, from automated high-throughput sequencing instruments to recombinant-DNA vectors that can convey color-coded genetic modifications in the chromosomes of live adult animals. This review covers the highlights of these advances, both theoretical and technological, along with a brief window into the promising science ahead. This article is part of the 60th Anniversary special issue.
Collapse
Affiliation(s)
- Steven W Barger
- Department of Geriatrics, Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA. .,Geriatric Research Education and Clinical Center, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA.
| |
Collapse
|
13
|
Christensen PC, Welch NC, Brideau C, Stys PK. Functional ionotropic glutamate receptors on peripheral axons and myelin. Muscle Nerve 2016; 54:451-9. [DOI: 10.1002/mus.25078] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 01/26/2016] [Accepted: 02/10/2016] [Indexed: 01/22/2023]
Affiliation(s)
- Pia Crone Christensen
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, 3330 Hospital Drive NW, University of Calgary; Calgary Alberta Canada T2N 4N1
| | - Nicole Cheryl Welch
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, 3330 Hospital Drive NW, University of Calgary; Calgary Alberta Canada T2N 4N1
| | - Craig Brideau
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, 3330 Hospital Drive NW, University of Calgary; Calgary Alberta Canada T2N 4N1
| | - Peter K. Stys
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, 3330 Hospital Drive NW, University of Calgary; Calgary Alberta Canada T2N 4N1
| |
Collapse
|
14
|
Abstract
White matter of the brain and spinal cord is irreversibly damaged by ischemia and trauma. Recent evidence indicates that despite the absence of synaptic elements, excitotoxic mechanisms play an important role in the pathogenesis of white matter damage. Glial cells, including astrocytes and oligodendrocytes, possess non-NMDA glutamate receptors and are injured by excessive exposure to AMPA/kainate agonists. In addition, the myelin sheath itself appears to respond directly to glutamate stimulation via AMPA receptors, which may also lead to injury of this key constituent of myelinated axons. During white matter anoxia/ischemia or trauma, endogenous glutamate is released mainly from axoplasmic pools in a nonvesicular fashion through Na+-dependent glutamate transporters, stimulated to operate in the glutamate efflux mode by collapse of transmembrane ion gradients and depolarization. It appears that parallel mechanisms are triggered by injurious stimuli, involving reverse Na+-Ca2+ exchange and voltage-gated Ca2+ channels producing Ca2+ overload of the axon cylinder, whereas glutamate release with AMPA receptor overactivation causes Ca2+-dependent damage to the ensheathing myelin and sup-porting glia. The emerging complexity of white matter injury mechanisms requires a thorough understanding of the interrelated steps to optimize therapeutic design.
Collapse
Affiliation(s)
- Peter K. Stys
- Division of Neuroscience, Loeb Health Research Institute, Ottawa Hospital-Civic Campus, University of Ottawa, Ottawa, Ontario, Canada,
| | - Shuxin Li
- Division of Neuroscience, Loeb Health Research Institute, Ottawa Hospital-Civic Campus, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
15
|
Glutamate signalling: A multifaceted modulator of oligodendrocyte lineage cells in health and disease. Neuropharmacology 2016; 110:574-585. [PMID: 27346208 DOI: 10.1016/j.neuropharm.2016.06.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/27/2016] [Accepted: 06/16/2016] [Indexed: 01/10/2023]
Abstract
Myelin is essential for the mammalian brain to function efficiently. Whilst many factors have been associated with regulating the differentiation of oligodendroglia and myelination, glutamate signalling might be particularly important for learning-dependent myelination. The majority of myelinated projection neurons are glutamatergic. Oligodendrocyte precursor cells receive glutamatergic synaptic inputs from unmyelinated axons and oligodendrocyte lineage cells express glutamate receptors which enable them to monitor and respond to changes in neuronal activity. Yet, what role glutamate plays for oligodendroglia is not fully understood. Here, we review glutamate signalling and its effects on oligodendrocyte lineage cells, and myelination in health and disease. Furthermore, we discuss whether glutamate signalling between neurons and oligodendroglia might lay the foundation to activity-dependent white matter plasticity. This article is part of the Special Issue entitled 'Oligodendrocytes in Health and Disease'.
Collapse
|
16
|
Ransom BR, Goldberg MP, Arai K, Baltan S. White Matter Pathophysiology. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00009-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
17
|
Cofilin as a Promising Therapeutic Target for Ischemic and Hemorrhagic Stroke. Transl Stroke Res 2015; 7:33-41. [PMID: 26670926 DOI: 10.1007/s12975-015-0438-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 12/01/2015] [Accepted: 12/03/2015] [Indexed: 01/22/2023]
Abstract
Neurovascular unit (NVU) is considered as a conceptual framework for investigating the mechanisms as well as developing therapeutic targets for ischemic and hemorrhagic stroke. From a molecular perspective, oxidative stress, excitotoxicity, inflammation, and disruption of the blood brain barrier are broad pathophysiological frameworks on the basis on which potential therapeutic candidates for ischemic and hemorrhagic stroke could be discussed. Cofilin is a potent actin-binding protein that severs and depolymerizes actin filaments in order to generate the dynamics of the actin cytoskeleton. Although studies of the molecular mechanisms of cofilin-induced reorganization of the actin cytoskeleton have been ongoing for decades, the multicellular functions of cofilin and its regulation in different molecular pathways are expanding beyond its primary role in actin cytoskeleton. This review focuses on the role of cofilin in oxidative stress, excitotoxicity, inflammation, and disruption of the blood brain barrier in the context of NVU as well as how and why cofilin could be studied further as a potential target for ischemic and hemorrhagic stroke.
Collapse
|
18
|
Abstract
Therapeutic repair of myelin disorders may be limited by the relatively slow rate of human oligodendrocyte differentiation. To identify appropriate pharmacological targets with which to accelerate differentiation of human oligodendrocyte progenitors (hOPCs) directly, we used CD140a/O4-based FACS of human forebrain and microarray to hOPC-specific receptors. Among these, we identified CHRM3, a M3R muscarinic acetylcholine receptor, as being restricted to oligodendrocyte-biased CD140a(+)O4(+) cells. Muscarinic agonist treatment of hOPCs resulted in a specific and dose-dependent blockade of oligodendrocyte commitment. Conversely, when hOPCs were cocultured with human neurons, M3R antagonist treatment stimulated oligodendrocytic differentiation. Systemic treatment with solifenacin, an FDA-approved muscarinic receptor antagonist, increased oligodendrocyte differentiation of transplanted hOPCs in hypomyelinated shiverer/rag2 brain. Importantly, solifenacin treatment of engrafted animals reduced auditory brainstem response interpeak latency, indicative of increased conduction velocity and thereby enhanced functional repair. Therefore, solifenacin and other selective muscarinic antagonists represent new adjunct approaches to accelerate repair by engrafted human progenitors.
Collapse
|
19
|
Titomanlio L, Fernández-López D, Manganozzi L, Moretti R, Vexler ZS, Gressens P. Pathophysiology and neuroprotection of global and focal perinatal brain injury: lessons from animal models. Pediatr Neurol 2015; 52:566-584. [PMID: 26002050 PMCID: PMC4720385 DOI: 10.1016/j.pediatrneurol.2015.01.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 01/16/2015] [Accepted: 01/24/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND Arterial ischemic stroke occurs more frequently in term newborns than in the elderly, and brain immaturity affects mechanisms of ischemic injury and recovery. The susceptibility to injury of the brain was assumed to be lower in the perinatal period as compared with childhood. This concept was recently challenged by clinical studies showing marked motor disabilities after stroke in neonates, with the severity of motor and cortical sensory deficits similar in both perinatal and childhood ischemic stroke. Our understanding of the triggers and the pathophysiological mechanisms of perinatal stroke has greatly improved in recent years, but many factors remain incompletely understood. METHODS In this review, we focus on the pathophysiology of perinatal stroke and on therapeutic strategies that can protect the immature brain from the consequences of stroke by targeting inflammation and brain microenvironment. RESULTS Studies in neonatal rodent models of cerebral ischemia have suggested a potential role for soluble inflammatory molecules as important modulators of injury and recovery. A great effort is underway to investigate neuroprotective molecules based on our increasing understanding of the pathophysiology. CONCLUSION In this review, we provide a comprehensive summary of new insights concerning pathophysiology of focal and global perinatal brain injury and their implications for new therapeutic approaches.
Collapse
Affiliation(s)
- Luigi Titomanlio
- Pediatric Emergency Department, APHP, Robert Debré Hospital, Paris, France
- Inserm, U1141, F-75019 Paris, France
| | - David Fernández-López
- Department of Neurology, University of California San Francisco, San Francisco, CA, 94158-0663, USA
| | - Lucilla Manganozzi
- Pediatric Emergency Department, APHP, Robert Debré Hospital, Paris, France
- Inserm, U1141, F-75019 Paris, France
| | | | - Zinaida S. Vexler
- Department of Neurology, University of California San Francisco, San Francisco, CA, 94158-0663, USA
| | - Pierre Gressens
- Inserm, U1141, F-75019 Paris, France
- Univ Paris Diderot, Sorbonne Paris Cité, UMRS 676, F-75019 Paris, France
- PremUP, Paris, France
- Centre for the Developing Brain, King’s College, St Thomas’ Campus, London SE1 7EH, UK
| |
Collapse
|
20
|
Kritis AA, Stamoula EG, Paniskaki KA, Vavilis TD. Researching glutamate - induced cytotoxicity in different cell lines: a comparative/collective analysis/study. Front Cell Neurosci 2015; 9:91. [PMID: 25852482 PMCID: PMC4362409 DOI: 10.3389/fncel.2015.00091] [Citation(s) in RCA: 259] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 02/26/2015] [Indexed: 12/21/2022] Open
Abstract
Although glutamate is one of the most important excitatory neurotransmitters of the central nervous system, its excessive extracellular concentration leads to uncontrolled continuous depolarization of neurons, a toxic process called, excitotoxicity. In excitotoxicity glutamate triggers the rise of intracellular Ca2+ levels, followed by up regulation of nNOS, dysfunction of mitochondria, ROS production, ER stress, and release of lysosomal enzymes. Excessive calcium concentration is the key mediator of glutamate toxicity through over activation of ionotropic and metabotropic receptors. In addition, glutamate accumulation can also inhibit cystine (CySS) uptake by reversing the action of the CySS/glutamate antiporter. Reversal of the antiporter action reinforces the aforementioned events by depleting neurons of cysteine and eventually glutathione’s reducing potential. Various cell lines have been employed in the pursuit to understand the mechanism(s) by which excitotoxicity affects the cells leading them ultimately to their demise. In some cell lines glutamate toxicity is exerted mainly through over activation of NMDA, AMPA, or kainate receptors whereas in other cell lines lacking such receptors, the toxicity is due to glutamate induced oxidative stress. However, in the greatest majority of the cell lines ionotropic glutamate receptors are present, co-existing to CySS/glutamate antiporters and metabotropic glutamate receptors, supporting the assumption that excitotoxicity effect in these cells is accumulative. Different cell lines differ in their responses when exposed to glutamate. In this review article the responses of PC12, SH-SY5Y, HT-22, NT-2, OLCs, C6, primary rat cortical neurons, RGC-5, and SCN2.2 cell systems are systematically collected and analyzed.
Collapse
Affiliation(s)
- Aristeidis A Kritis
- Laboratory of Physiology, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki Greece
| | - Eleni G Stamoula
- Laboratory of Physiology, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki Greece
| | - Krystallenia A Paniskaki
- Laboratory of Physiology, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki Greece
| | - Theofanis D Vavilis
- Laboratory of Physiology, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki Greece
| |
Collapse
|
21
|
Back SA, Rosenberg PA. Pathophysiology of glia in perinatal white matter injury. Glia 2014; 62:1790-815. [PMID: 24687630 DOI: 10.1002/glia.22658] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/13/2014] [Accepted: 02/27/2014] [Indexed: 12/12/2022]
Abstract
Injury to the preterm brain has a particular predilection for cerebral white matter. White matter injury (WMI) is the most common cause of brain injury in preterm infants and a major cause of chronic neurological morbidity including cerebral palsy. Factors that predispose to WMI include cerebral oxygenation disturbances and maternal-fetal infection. During the acute phase of WMI, pronounced oxidative damage occurs that targets late oligodendrocyte progenitors (pre-OLs). The developmental predilection for WMI to occur during prematurity appears to be related to both the timing of appearance and regional distribution of susceptible pre-OLs that are vulnerable to a variety of chemical mediators including reactive oxygen species, glutamate, cytokines, and adenosine. During the chronic phase of WMI, the white matter displays abberant regeneration and repair responses. Early OL progenitors respond to WMI with a rapid robust proliferative response that results in a several fold regeneration of pre-OLs that fail to terminally differentiate along their normal developmental time course. Pre-OL maturation arrest appears to be related in part to inhibitory factors that derive from reactive astrocytes in chronic lesions. Recent high field magnetic resonance imaging (MRI) data support that three distinct forms of chronic WMI exist, each of which displays unique MRI and histopathological features. These findings suggest the possibility that therapies directed at myelin regeneration and repair could be initiated early after WMI and monitored over time. These new mechanisms of acute and chronic WMI provide access to a variety of new strategies to prevent or promote repair of WMI in premature infants.
Collapse
Affiliation(s)
- Stephen A Back
- Department of Pediatrics, Oregon Health and Science University, Portland, Oregon; Department of Neurology, Oregon Health and Science University, Portland, Oregon
| | | |
Collapse
|
22
|
Hossain S, Liu HN, Fragoso G, Almazan G. Agonist-induced down-regulation of AMPA receptors in oligodendrocyte progenitors. Neuropharmacology 2014; 79:506-14. [PMID: 24412648 DOI: 10.1016/j.neuropharm.2013.12.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 12/09/2013] [Accepted: 12/23/2013] [Indexed: 10/25/2022]
Abstract
Prolonged exposure of oligodendrocyte progenitor cultures to non-toxic concentrations of glutamate receptor agonists for 24 h decreased cellular proliferation mediated by α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Since prolonged agonist stimulation can regulate the expression of various families of receptors, we examined this possibility. Pretreatment of progenitor cultures with 100 μM kainic acid (KA) for 1-24 h caused a time-dependent decrease in AMPA receptor activity, determined by agonist-induced (45)Ca(2+) uptake. The maximum effect (70-80% decrease), observed in the 24 h-pretreated cells, was accompanied by a significant reduction in AMPA receptor subunits, as determined by Western blotting. GluR2/3 and GluR4 subunits were the most affected. Receptor down-regulation and (45)Ca(2+) uptake were only partially reversible upon KA removal. Furthermore, 24 h co-treatment of cultures with CNQX blocked the KA-induced decreases in calcium uptake. To address whether calpain, a calcium-activated protease, was implicated in the regulation of the AMPA receptor subunits, cultures were treated with the specific inhibitor PD150606 alone or in combination with KA for 24 h. Calpain inhibition significantly increased GluR1 in both conditions and partly reversed downregulation of GluR4 by KA. Collectively, these results indicate that calpain is not involved in the agonist-induced down-regulation of AMPA receptors subunits 2/3 in oligodendrocyte progenitors, while it downregulates GluR1 and GluR4.
Collapse
Affiliation(s)
- Shireen Hossain
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Hsueh-Ning Liu
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Gabriela Fragoso
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Guillermina Almazan
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
23
|
Simonishvili S, Jain M, Li H, Levison S, Wood T. Identification of Bax-interacting proteins in oligodendrocyte progenitors during glutamate excitotoxicity and perinatal hypoxia-ischemia. ASN Neuro 2013; 5:e00131. [PMID: 24195677 PMCID: PMC3891358 DOI: 10.1042/an20130027] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
OPC (oligodendrocyte progenitor cell) death contributes significantly to the pathology and functional deficits following hypoxic-ischemic injury in the immature brain and to deficits resulting from demyelinating diseases, trauma and degenerative disorders in the adult CNS. Glutamate toxicity is a major cause of oligodendroglial death in diverse CNS disorders, and previous studies have demonstrated that AMPA/kainate receptors require the pro-apoptotic protein Bax in OPCs undergoing apoptosis. The goal of the present study was to define the pro-apoptotic and anti-apoptotic effectors that regulate Bax in healthy OPCs and after exposure to excess glutamate in vitro and following H-I (hypoxia-ischemia) in the immature rat brain. We show that Bax associates with a truncated form of Bid, a BH3-only domain protein, subsequent to glutamate treatment. Furthermore, glutamate exposure reduces Bax association with the anti-apoptotic Bcl family member, Bcl-xL. Cell fractionation studies demonstrated that both Bax and Bid translocate from the cytoplasm to mitochondria during the early stages of cell death consistent with a role for Bid as an activator, whereas Bcl-xL, which normally complexes with both Bax and Bid, disassociates from these complexes when OPCs are exposed to excess glutamate. Bax remained unactivated in the presence of insulin-like growth factor-1, and the Bcl-xL complexes were protected. Our data similarly demonstrate loss of Bcl-xL-Bax association in white matter following H-I and implicate active Bad in Bax-mediated OPC death. To identify other Bax-binding partners, we used proteomics and identified cofilin as a Bax-associated protein in OPCs. Cofilin and Bax associated in healthy OPCs, whereas the Bax-cofilin association was disrupted during glutamate-induced OPC apoptosis.
Collapse
Key Words
- apoptosis
- bcl-xl
- bid
- cofilin
- insulin-like growth factor 1 (igf-i)
- oligodendrocyte
- acn, acetonitrile
- adf, actin depolymerizing factor
- af488, alexa fluor 488
- af546, alexa fluor 546
- cca, common carotid artery
- cl, contralateral
- cns, central nervous system
- dmem, dulbecco’s modified eagle’s medium
- fbs, fetal bovine serum
- fgf-2, fibroblast growth factor-2
- h–i, hypoxia–ischemia
- igf, insulin-like growth factor
- il, ipsilateral
- ip, immunoprecipitation
- mem, minimal essential media
- opc, oligodendrocyte progenitor cell
- pic, protease inhibitor cocktail
- tbid, truncated bid
- vdac, voltage-dependent anion channel
Collapse
Affiliation(s)
- Sopio Simonishvili
- *Department of Neurology & Neuroscience, New Jersey Medical School Cancer Center, Rutgers Biomedical & Health Sciences, Newark, NJ 07101, U.S.A
| | - Mohit Raja Jain
- †Center for Advanced Proteomic Research and Department of Biochemistry and Molecular Biology, New Jersey Medical School Cancer Center, Rutgers Biomedical & Health Sciences, Newark, NJ 07101, U.S.A
| | - Hong Li
- †Center for Advanced Proteomic Research and Department of Biochemistry and Molecular Biology, New Jersey Medical School Cancer Center, Rutgers Biomedical & Health Sciences, Newark, NJ 07101, U.S.A
| | - Steven W. Levison
- *Department of Neurology & Neuroscience, New Jersey Medical School Cancer Center, Rutgers Biomedical & Health Sciences, Newark, NJ 07101, U.S.A
| | - Teresa L. Wood
- *Department of Neurology & Neuroscience, New Jersey Medical School Cancer Center, Rutgers Biomedical & Health Sciences, Newark, NJ 07101, U.S.A
- 1To whom correspondence should be addressed (email )
| |
Collapse
|
24
|
Song M, Yu SP. Ionic regulation of cell volume changes and cell death after ischemic stroke. Transl Stroke Res 2013; 5:17-27. [PMID: 24323733 DOI: 10.1007/s12975-013-0314-x] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 10/16/2013] [Accepted: 11/19/2013] [Indexed: 12/20/2022]
Abstract
Stroke is a leading cause of human death and disability in the USA and around the world. Shortly after the cerebral ischemia, cell swelling is the earliest morphological change in injured neuronal, glial, and endothelial cells. Cytotoxic swelling directly results from increased Na(+) (with H2O) and Ca(2+) influx into cells via ionic mechanisms evoked by membrane depolarization and a number of harmful factors such as glutamate accumulation and the production of oxygen reactive species. During the sub-acute and chronic phases after ischemia, injured cells may show a phenotype of cell shrinkage due to complex processes involving membrane receptors/channels and programmed cell death signals. This review will introduce some progress in the understanding of the regulation of pathological cell volume changes and the involved receptors and channels, including NMDA and AMPA receptors, acid-sensing ion channels, hemichannels, transient receptor potential channels, and KCNQ channels. Moreover, accumulating evidence supports a key role of energy deficiency and dysfunction of Na(+)/K(+)-ATPase in ischemia-induced cell volume changes and cell death. Specifically, the Na(+) pump failure is a prerequisite for disruption of ionic homeostasis including a pro-apoptotic disruption of the K(+) homeostasis. Finally, we will introduce the concept of hybrid cell death as a result of the Na(+) pump failure in cultured cells and the ischemic brain. The goal of this review is to outline recent understanding of the ionic mechanism of ischemic cytotoxicity and suggest innovative ideas for future translational research.
Collapse
Affiliation(s)
- Mingke Song
- Department of Anesthesiology, Emory University School of Medicine, 101 Woodruff Circle, WMB Building Suite 620, Atlanta, GA, 30322, USA
| | | |
Collapse
|
25
|
Neurotransmitter-triggered transfer of exosomes mediates oligodendrocyte-neuron communication. PLoS Biol 2013; 11:e1001604. [PMID: 23874151 PMCID: PMC3706306 DOI: 10.1371/journal.pbio.1001604] [Citation(s) in RCA: 651] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 05/29/2013] [Indexed: 11/23/2022] Open
Abstract
Neuronal activity provokes myelinating oligodendrocytes to release exosomes by stimulation of ionotropic glutamate receptors, and that once released, these vesicles are internalized by neurons conveying neuroprotection. Reciprocal interactions between neurons and oligodendrocytes are not only crucial for myelination, but also for long-term survival of axons. Degeneration of axons occurs in several human myelin diseases, however the molecular mechanisms of axon-glia communication maintaining axon integrity are poorly understood. Here, we describe the signal-mediated transfer of exosomes from oligodendrocytes to neurons. These endosome-derived vesicles are secreted by oligodendrocytes and carry specific protein and RNA cargo. We show that activity-dependent release of the neurotransmitter glutamate triggers oligodendroglial exosome secretion mediated by Ca2+ entry through oligodendroglial NMDA and AMPA receptors. In turn, neurons internalize the released exosomes by endocytosis. Injection of oligodendroglia-derived exosomes into the mouse brain results in functional retrieval of exosome cargo in neurons. Supply of cultured neurons with oligodendroglial exosomes improves neuronal viability under conditions of cell stress. These findings indicate that oligodendroglial exosomes participate in a novel mode of bidirectional neuron-glia communication contributing to neuronal integrity. Brain function largely depends on the communication between electrically excitable neurons and surrounding glial cells. Myelinating oligodendrocytes are a type of brain cell that insulate major neuronal processes (axons) and help to sustainably maintain axonal health, which is poorly understood in molecular terms. Several cell types release microvesicles termed exosomes that include genetic information (primarily RNA) and can act as vehicles transferring specific cargo to target cells. Here, we demonstrate that exosomes secreted by oligodendrocytes in response to neuronal signals enter neurons to make their cargo functionally available to the neuronal metabolism. We revealed in cultured cells that exosome release from oligodendrocytes is triggered by the neurotransmitter glutamate through activation of ionotropic glutamate receptors. We also show that glial exosomes are internalized by neurons via an endocytic pathway. By modifying oligodendroglial exosomes with a reporter enzyme, we could demonstrate that the exosome cargo is recovered by target neurons in culture as well as in vivo after injection of exosomes into the mouse brain. Neurons challenged with stressful growth conditions were protected when treated with oligodendroglial exosomes. The study introduces a new concept of reciprocal cell communication in the nervous system and identifies the signal-mediated transfer of exosomes from oligodendrocytes to neurons contributing to the preservation of axonal health.
Collapse
|
26
|
Benjamins JA, Nedelkoska L, Bealmear B, Lisak RP. ACTH protects mature oligodendroglia from excitotoxic and inflammation-related damage in vitro. Glia 2013; 61:1206-17. [PMID: 23832579 DOI: 10.1002/glia.22504] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 03/07/2013] [Indexed: 12/20/2022]
Abstract
Corticosteroids (CS) are widely employed to treat relapses in multiple sclerosis (MS). Endogenous ACTH is a 39-amino acid peptide that, among other functions, stimulates CS production. Exogenous ACTH 1-39 is used to treat MS relapses, presumably by stimulating endogenous CS production. However, unlike CS, ACTH binds to melanocortin receptors, found in the central nervous system (CNS) as well as on inflammatory cells. Since glia are implicated in MS and other neurodegenerative diseases, and oligodendroglia (OL) are more sensitive to injury than other glia, we characterized the protective effects of ACTH on OL in vitro without the confounding effects of CS. Rat brain cultures containing OL, astrocytes (AS), and microglia (MG) were incubated for 1 day with potentially cytotoxic agents with or without preincubation with ACTH 1-39. The cytotoxic agents killed 55-70% of mature OL, but caused little or no death of AS or MG at the concentrations used. ACTH protected OL from death induced by staurosporine, AMPA, NMDA, kainate, quinolinic acid, or reactive oxygen species, but did not protect against kynurenic acid or nitric oxide. The protective effects of ACTH were dose dependent, and decreased OL death induced by the different agents by 30-60% at 200 nM ACTH. We show for the first time that melanocortin 4 receptor is expressed on OL in addition to MG and AS. In summary, ACTH 1-39 protects OL in vitro from several excitotoxic and inflammation-related insults. ACTH may be activating melanocortin receptors on OL or alternately on AS or MG to prevent OL death.
Collapse
Affiliation(s)
- Joyce A Benjamins
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | | | | | | |
Collapse
|
27
|
Molecular pathology, classification, and diagnosis of sporadic human prion disease variants. Folia Neuropathol 2012; 4:AN20110031. [PMID: 22356284 PMCID: PMC3284768 DOI: 10.1042/an20110031] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Glutamate excitotoxicity is a major pathogenic process implicated in many neurodegenerative conditions, including AD (Alzheimer's disease) and following traumatic brain injury. Occurring predominantly from over-stimulation of ionotropic glutamate receptors located along dendrites, excitotoxic axonal degeneration may also occur in white matter tracts. Recent identification of axonal glutamate receptor subunits within axonal nanocomplexes raises the possibility of direct excitotoxic effects on axons. Individual neuronal responses to excitotoxicity are highly dependent on the complement of glutamate receptors expressed by the cell, and the localization of the functional receptors. To enable isolation of distal axons and targeted excitotoxicity, murine cortical neuron cultures were prepared in compartmented microfluidic devices, such that distal axons were isolated from neuronal cell bodies. Within the compartmented culture system, cortical neurons developed to relative maturity at 11 DIV (days in vitro) as demonstrated by the formation of dendritic spines and clustering of the presynaptic protein synaptophysin. The isolated distal axons retained growth cone structures in the absence of synaptic targets, and expressed glutamate receptor subunits. Glutamate treatment (100 μM) to the cell body chamber resulted in widespread degeneration within this chamber and degeneration of distal axons in the other chamber. Glutamate application to the distal axon chamber triggered a lesser degree of axonal degeneration without degenerative changes in the untreated somal chamber. These data indicate that in addition to current mechanisms of indirect axonal excitotoxicity, the distal axon may be a primary target for excitotoxicity in neurodegenerative conditions.
Collapse
|
28
|
Morales I, Rodriguez M. Self-induced accumulation of glutamate in striatal astrocytes and basal ganglia excitotoxicity. Glia 2012; 60:1481-94. [PMID: 22715058 DOI: 10.1002/glia.22368] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Revised: 05/10/2012] [Accepted: 05/21/2012] [Indexed: 01/18/2023]
Abstract
Excitotoxicity induced by high levels of extracellular glutamate (GLU) has been proposed as a cause of cell degeneration in basal ganglia disorders. This phenomenon is normally prevented by the astrocytic GLU-uptake and the GLU-catabolization to less dangerous molecules. However, high-GLU can induce reactive gliosis which could change the neuroprotective role of astrocytes. The striatal astrocyte response to high GLU was studied here in an in vivo rat preparation. The transient striatal perfusion of GLU (1 h) by reverse microdialysis induced complex reactive gliosis which persisted for weeks and which was different for radial-like glia, protoplasmic astrocytes and fibrous astrocytes. This gliosis was accompanied by a persistent cytosolic accumulation of GLU (immunofluorescence quantified by confocal microscope), which persisted for weeks (self-induced glutamate accumulation), and which was associated to a selective decrease of glutamine synthetase activity. This massive and persistent self-induced glutamate accumulation in striatal astrocytes could be an additional factor for the GLU-induced excitotoxicity, which has been implicated in the progression of different basal ganglia disorders.
Collapse
Affiliation(s)
- Ingrid Morales
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La Laguna, La Laguna, Tenerife, Canary Islands, Spain
| | | |
Collapse
|
29
|
Kaur C, Rathnasamy G, Ling EA. Roles of activated microglia in hypoxia induced neuroinflammation in the developing brain and the retina. J Neuroimmune Pharmacol 2012; 8:66-78. [PMID: 22367679 DOI: 10.1007/s11481-012-9347-2] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 02/09/2012] [Indexed: 12/14/2022]
Abstract
Amoeboid microglial cells (AMCs) in the developing brain display surface receptors and antigens shared by the monocyte-derived tissue macrophages. Activation of AMCs in the perinatal brain has been associated with periventricular white matter damage in hypoxic-ischemic conditions. The periventricular white matter, where the AMCs preponderate, is selectively vulnerable to hypoxia as manifested by death of premyelinating oligodendrocytes and degeneration of axons leading to neonatal mortality and long-term neurodevelopmental deficits. AMCs respond vigorously to hypoxia by producing excess amounts of inflammatory cytokines e.g. the tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) along with glutamate, nitric oxide (NO) and reactive oxygen species which collectively cause oligodendrocyte death, axonal degeneration as well as disruption of the immature blood brain barrier. A similar phenomenon is observed in the hypoxic developing cerebellum in which activated AMCs induced Purkinje neuronal death through production of TNF-α and IL-1β via their respective receptors. Hypoxia is also implicated in retinopathy of prematurity in which activation of AMCs has been shown to cause retinal ganglion cell death through production of TNF-α and IL-1β and NO. Because AMCs play a pivotal role in hypoxic injuries in the developing brain affecting both neurons and oligodendrocytes, a fuller understanding of the underlying molecular mechanisms of microglial activation under such conditions would be desirable for designing of a novel therapeutic strategy for management of hypoxic damage.
Collapse
Affiliation(s)
- Charanjit Kaur
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, 4 Medical Drive, MD10, Singapore 117597, Singapore.
| | | | | |
Collapse
|
30
|
Sekler I, Silverman WF. Zinc homeostasis and signaling in glia. Glia 2012; 60:843-50. [DOI: 10.1002/glia.22286] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 12/02/2011] [Indexed: 11/08/2022]
|
31
|
Griesmaier E, Keller M. Glutamate receptors — Prenatal insults, long-term consequences. Pharmacol Biochem Behav 2012; 100:835-40. [DOI: 10.1016/j.pbb.2011.04.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 03/23/2011] [Accepted: 04/15/2011] [Indexed: 10/18/2022]
|
32
|
Abstract
Glutamate toxicity from hypoxia-ischaemia during the perinatal period causes white matter injury that can result in long-term motor and intellectual disability. Blocking ionotropic glutamate receptors (GluRs) has been shown to inhibit oligodendrocyte injury in vitro, but GluR antagonists have not yet proven helpful in clinical studies. The opposite approach of activating GluRs on developing oligodendrocytes shows promise in experimental studies on rodents as reported by Jartzie et al., in this issue. Group I metabotropic glutamate receptors (mGluRs) are expressed transiently on developing oligodendrocytes in humans during the perinatal period, and the blood-brain-barrier permeable agonist of group I mGluRs, 1-aminocyclopentane-trans-1,3-dicarboxylic acid (ACPD), reduces white matter damage significantly in a rat model of perinatal hypoxia-ischaemia. The results suggest drugs activating this class of GluRs could provide a new therapeutic approach for preventing cerebral palsy and other neurological consequences of diffuse white matter injury in premature infants.
Collapse
|
33
|
Molecular Pathophysiology of White Matter Anoxic-Ischemic Injury. Stroke 2011. [DOI: 10.1016/b978-1-4160-5478-8.10008-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
34
|
Kuzhandaivel A, Nistri A, Mladinic M. Kainate-mediated excitotoxicity induces neuronal death in the rat spinal cord in vitro via a PARP-1 dependent cell death pathway (Parthanatos). Cell Mol Neurobiol 2010; 30:1001-12. [PMID: 20502958 PMCID: PMC11498824 DOI: 10.1007/s10571-010-9531-y] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 05/11/2010] [Indexed: 01/14/2023]
Abstract
Kainate is an effective excitotoxic agent to lesion spinal cord networks, thus providing an interesting model for investigating basic mechanisms of spinal cord injury. The present study aimed at revealing the type and timecourse of cell death in rat neonatal spinal cord preparations in vitro exposed to 1 h excitotoxic insult with kainate. Substantial numbers of neurons rather than glia showed pyknosis (albeit without necrosis and with minimal apoptosis occurrence) already apparent on kainate washout and peaking 12 h later with dissimilar spinal topography. Neurons appeared to suffer chiefly through a process involving anucleolytic pyknosis mediated by strong activation of poly(ADP-ribose)polymerase-1 (PARP-1) that generated poly ADP-ribose and led to nuclear translocation of the apoptotic inducing factor (AIF) with DNA damage. This process had the hallmarks of parthanatos-type neuronal death. The PARP-1 inhibitor 6-5(H)-phenathridione applied immediately after kainate washout significantly prevented pyknosis in a dose-dependent fashion and inhibited PARP-1-dependent nuclear AIF translocation. Conversely, the caspase-3 inhibitor II was ineffective against neuronal damage. Our results suggest that excitotoxicity of spinal networks was mainly directed to neurons and mediated by PARP-1 death pathways, indicating this mechanism as a potential target for neuroprotection to limit the acute damage to the local circuitry.
Collapse
Affiliation(s)
- Anujaianthi Kuzhandaivel
- Neurobiology Sector, International School for Advanced Studies (SISSA), Via Bonomea 265, 34136 Trieste, Italy
| | - Andrea Nistri
- Neurobiology Sector, International School for Advanced Studies (SISSA), Via Bonomea 265, 34136 Trieste, Italy
- SPINAL (Spinal Person Injury Neurorehabilitation Applied Laboratory), Istituto di Medicina Fisica e Riabilitazione, Via Gervasutta 48, 33100 Udine, Italy
| | - Miranda Mladinic
- Neurobiology Sector, International School for Advanced Studies (SISSA), Via Bonomea 265, 34136 Trieste, Italy
- SPINAL (Spinal Person Injury Neurorehabilitation Applied Laboratory), Istituto di Medicina Fisica e Riabilitazione, Via Gervasutta 48, 33100 Udine, Italy
| |
Collapse
|
35
|
Nandhu M, Paul J, Mathew J, Peeyush Kumar T, Paulose C. GYKI-52466: A potential therapeutic agent for glutamate-mediated excitotoxic injury in Cerebral Palsy. Med Hypotheses 2010; 74:619-20. [DOI: 10.1016/j.mehy.2009.10.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2009] [Revised: 10/16/2009] [Accepted: 10/18/2009] [Indexed: 10/20/2022]
|
36
|
Pitt D, Gonzales E, Cross AH, Goldberg MP. Dysmyelinated axons in shiverer mice are highly vulnerable to alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptor-mediated toxicity. Brain Res 2009; 1309:146-54. [PMID: 19896473 DOI: 10.1016/j.brainres.2009.10.066] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2009] [Revised: 10/25/2009] [Accepted: 10/27/2009] [Indexed: 01/01/2023]
Abstract
Glutamate excitotoxicity plays a role in white matter injury in many neurological diseases. Oligodendrocytes in particular are highly vulnerable to excitotoxicity, mediated through activation of AMPA/kainate receptors. Myelin may also be injured independently via NMDA (N-methyl-D-aspartic acid) receptors located on peripheral oligodendroglial processes. Central axons are susceptible to glutamate receptor activation in vivo, but it is unclear whether this is mediated directly by activation of receptors expressed on axons, or indirectly through glutamate toxicity of myelin or neighboring glial cells. We examined axonal vulnerability in mice deficient in myelin basic protein (shiverer), also expressing yellow fluorescent protein (YFP) in a subset of axons. YFP fluorescence, EM, and mouse behavior were assessed 24 h after microstereotactical injections of S-AMPA or NMDA into lumbar dorsal columns. S-AMPA injection led to impaired rotarod performance and widespread axonal degeneration and was more pronounced in shiverer mice than controls. In contrast, NMDA injection did not cause axonal injury or behavioral changes in either group. These results indicate that spinal cord axons in vivo are vulnerable to toxicity mediated by AMPA but not NMDA receptors. The presence of compact myelin is not required for excitotoxic axon damage, and its absence may increase vulnerability. Further understanding of AMPA receptor-mediated axonal toxicity may provide new targets for neuroprotective therapy in WM diseases.
Collapse
Affiliation(s)
- David Pitt
- Department of Neurology, Division of Neuroimmunology, The Ohio State University, 460 W. 12th Ave., Columbus, OH 43210, USA.
| | | | | | | |
Collapse
|
37
|
Griesmaier E, Keller M. Neuroprotective strategies in excitotoxic brain injury: potential applications to the preterm brain. FUTURE NEUROLOGY 2009. [DOI: 10.2217/fnl.09.28] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Neuronal and oligodendroglial cell death owing to increased glutamate levels plays an important role in the pathophysiology of hypoxic-, ischemic- and inflammation-mediated brain injury as well as in disorders such as epilepsy, Alzheimer’s, Parkinson’s or Huntington’s disease. In addition, excitotoxic brain injury is known to be a major contributing factor to brain injury in preterm infants. Excitotoxicity is characterized as excessive glutamatergic activation of postsynaptic receptors that consequently leads to cell injury and cell death. The major excitatory amino acid neurotransmitter is glutamate. Glutamate plays a key role in brain development, affecting progenitor cell differentiation, proliferation, migration and survival. In physiological conditions the presence of glutamate in the synapse is regulated by ATP-dependent glutamate transporters in neurons and glial cells, with astrocytes being responsible for a major part of glutamate uptake in the brain. In pathologic circumstances the function of the transporters is impaired, leading to glutamate accumulation in the synaptic cleft and in turn excessive activation of postsynaptic glutamate receptors with subsequent massive Ca2+ influx, activation of neuronal nitric oxide synthase, translocation of proapoptotic genes to the mitochondria, mitochondrial dysfunction, release of cytochrome C into the cytosol, activation of caspases and subsequent cell death. Based on the pathogenic concept of an overactivation of the excitatory pathways, glutamate receptors have been a longstanding therapeutic target for rational drug design. This article reviews the pathophysiology of excitotoxic brain injury in the example of preterm brain injury, as well as current research on therapeutic antiexcitotoxic strategies.
Collapse
Affiliation(s)
- Elke Griesmaier
- Department of Pediatrics IV, Medical University Innsbruck, Austria, Anichstr. 35, 6020 Innsbruck, Austria
| | - Matthias Keller
- Department of Pediatrics I University Hospital Essen, Hufelandstraße 55, 45147 Essen Germany
| |
Collapse
|
38
|
DeSilva TM, Kabakov AY, Goldhoff PE, Volpe JJ, Rosenberg PA. Regulation of glutamate transport in developing rat oligodendrocytes. J Neurosci 2009; 29:7898-908. [PMID: 19535601 PMCID: PMC2926807 DOI: 10.1523/jneurosci.6129-08.2009] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Revised: 03/16/2009] [Accepted: 04/08/2009] [Indexed: 12/22/2022] Open
Abstract
Glutamate released from synaptic vesicles mediates excitatory neurotransmission by stimulating glutamate receptors. Glutamate transporters maintain low synaptic glutamate levels critical for this process, a role primarily attributed to astrocytes. Recently, vesicular release of glutamate from unmyelinated axons in the rat corpus callosum has been shown to elicit AMPA receptor-mediated currents in glial progenitor cells. Glutamate transporters are the only mechanism of glutamate clearance, yet very little is known about the role of glutamate transporters in normal development of oligodendrocytes (OLs) or in excitotoxic injury to OLs. We found that OLs in culture are capable of sodium-dependent glutamate uptake with a K(m) of 10 +/- 2 microm and a V(max) of 2.6, 5.0, and 3.8 nmol x min(-1) x mg(-1) for preoligodendrocytes, immature, and mature OLs, respectively. Surprisingly, EAAC1, thought to be exclusively a neuronal transporter, contributes more to [(3)H]l-glutamate uptake in OLs than GLT1 or GLAST. These data suggest that glutamate transporters on oligodendrocytes may serve a critical role in maintaining glutamate homeostasis at a time when unmyelinated callosal axons are engaging in glutamatergic signaling with glial progenitors. Furthermore, GLT1 was significantly increased in cultured mature OLs contrary to in vivo data in which we have shown that, although GLT1 is present on developing OLs when unmyelinated axons are prevalent in the developing rat corpus callosum, after myelination, GLT1 is not expressed on mature OLs. The absence of GLT1 in mature OLs in the rat corpus callosum and its presence in mature rat cultured OLs may indicate that a signaling process in vivo is not activated in vitro.
Collapse
Affiliation(s)
- Tara M. DeSilva
- Department of Neurology and F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Anatoli Y. Kabakov
- Department of Neurology and F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Patricia E. Goldhoff
- Department of Neurology and F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Joseph J. Volpe
- Department of Neurology and F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Paul A. Rosenberg
- Department of Neurology and F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
39
|
Brighina E, Bresolin N, Pardi G, Rango M. Human fetal brain chemistry as detected by proton magnetic resonance spectroscopy. Pediatr Neurol 2009; 40:327-42. [PMID: 19380068 DOI: 10.1016/j.pediatrneurol.2008.11.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2008] [Revised: 11/03/2008] [Accepted: 11/19/2008] [Indexed: 11/25/2022]
Abstract
Magnetic resonance spectroscopy represents an invaluable tool for the in vivo study of brain development at the chemistry level. Whereas magnetic resonance spectroscopy has received wide attention in pediatric and adult settings, only a few studies were performed on the human fetal brain. They revealed changes occurring throughout gestation in the levels of the main metabolites detected by proton magnetic resonance spectroscopy (N-acetylaspartate, choline, myo-inositol, creatine, and glutamate), providing a reference for the normal metabolic brain development. Throughout the third trimester of gestation, N-acetylaspartate gradually increases, whereas choline undergoes a slow reduction during the process of myelination. Less clear are the modifications in creatine, myo-inositol, and glutamate levels. Under conditions of fetal distress, the meaning of lactate detection is unclear, and further studies are needed. Another field for investigation involves the possibility of early detection of glutamate levels in fetuses at risk for hypoxic-ischemic encephalopathy, because the role of glutamate excitotoxicity in this context is well-established. Because metabolic modifications may precede functional or morphologic changes in the central nervous system, magnetic resonance spectroscopy may likely serve as a powerful, noninvasive tool for the early diagnosis and prognosis of different pathologic conditions.
Collapse
Affiliation(s)
- Erika Brighina
- Foundation Instituto di Ricerca e Cura a Carattere Scientifico Policlinico, Mangiagalli and Regina Elena, University of Milan, Milan, Italy
| | | | | | | |
Collapse
|
40
|
Regulated release of BDNF by cortical oligodendrocytes is mediated through metabotropic glutamate receptors and the PLC pathway. ASN Neuro 2009; 1:AN20090006. [PMID: 19570026 PMCID: PMC2695578 DOI: 10.1042/an20090006] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
A number of studies suggest that OLGs (oligodendrocytes), the myelinating cells of the central nervous system, are also a source of trophic molecules, such as neurotrophins that may influence survival of proximate neurons. What is less clear is how the release of these molecules may be regulated. The present study investigated the effects of BDNF (brain-derived neurotrophic factor) derived from cortical OLGs on proximate neurons, as well as regulatory mechanisms mediating BDNF release. Initial work determined that BDNF derived from cortical OLGs increased the numbers of VGLUT1 (vesicular glutamate transporter 1)-positive glutamatergic cortical neurons. Furthermore, glutamate acting through metabotropic, and not AMPA/kainate or NMDA (N-methyl-d-aspartate), receptors increased BDNF release. The PLC (phospholipase C) pathway is a key mediator of metabotropic actions to release BDNF in astrocytes and neurons. Treatment of OLGs with the PLC activator m-3M3FBS [N-(3-trifluoromethylphenyl)-2,4,6-trimethylbenzenesulfonamide] induced robust release of BDNF. Moreover, release elicited by the metabotropic receptor agonist ACPD [trans-(1S,3R)-1-aminocyclopentane-1,3-dicarboxylic acid] was inhibited by the PLC antagonist U73122, the IP3 (inositol triphosphate 3) receptor inhibitor 2-APB (2-aminoethoxydiphenylborane) and the intracellular calcium chelator BAPTA/AM [1,2-bis-(o-aminophenoxy)ethane-N,N,N',N'-tetra-acetic acid tetrakis(acetoxymethyl ester)]. Taken together, these results suggest that OLG lineage cells release BDNF, a molecule trophic for proximate neurons. BDNF release is regulated by glutamate acting through mGluRs (metabotropic glutamate receptors) and the PLC pathway. Thus glutamate and BDNF may be molecules that support neuron-OLG interactions in the cortex.
Collapse
Key Words
- brain-derived neurotrophic factor (bdnf)
- metabotropic glutamate receptor
- oligodendrocyte
- phospholipase c
- acpd, trans-(1s,3r)-1-aminocyclopentane-1,3-dicarboxylic acid
- 2-apb, 2-aminoethoxydiphenylborane
- bapta/am, 1,2-bis-(o-aminophenoxy)ethane-n,n,n′,n′-tetra-acetic acid tetrakis(acetoxymethyl ester
- bdnf, brain-derived neurotrophic factor
- bf, basal forebrain
- cc, corpus callosal
- cns, central nervous system
- dcg-iv, (2s,2′r,3′r)-2-(2′,3′-dicarboxycyclopropyl)glycine
- dhpg, (rs)-3,5-dihydroxyphenylglycine
- dmso, dimethyl sulfoxide
- ecl, enhanced chemiluminescence
- gdnf, glial cell line-derived neurotrophic factor
- igf-1, insulin-like growth factor-1
- ip3, inositol trisphosphate
- mcpg, (s)-α-methyl-4-carboxyphenylglycine
- mem, minimal essential medium
- mglur, metabotropic glutamate receptor
- m-3m3fbs, n-(3-trifluoromethylphenyl)-2,4,6-trimethylbenzenesulfonamide
- nm-15, nutrient medium-15
- nmda, n-methyl-d-aspartate
- nsfm, neuron serum-free medium
- nt, neurotrophin
- ocm, oligodendrocyte-derived conditioned medium
- olg, oligodendrocyte
- osfm, olg serum-free medium
- plc, phospholipase c
- vamp2, vesicle-associated membrane protein 2
- vglut1, vesicular glutamate transporter 1
Collapse
|
41
|
Bakiri Y, Burzomato V, Frugier G, Hamilton NB, Káradóttir R, Attwell D. Glutamatergic signaling in the brain's white matter. Neuroscience 2009; 158:266-74. [PMID: 18314276 DOI: 10.1016/j.neuroscience.2008.01.015] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2007] [Accepted: 01/05/2008] [Indexed: 11/22/2022]
Abstract
Glutamatergic signaling has been exceptionally well characterized in the brain's gray matter, where it underlies fast information processing, learning and memory, and also generates the neuronal damage that occurs in pathological conditions such as stroke. The role of glutamatergic signaling in the white matter, an area until recently thought to be devoid of synapses, is less well understood. Here we review what is known, and highlight what is not known, of glutamatergic signaling in the white matter. We focus on how glutamate is released, the location and properties of the receptors it acts on, the interacting molecules that may regulate trafficking or signaling of the receptors, the possible functional roles of glutamate in the white matter, and its pathological effects including the possibility of treating white matter disorders with glutamate receptor blockers.
Collapse
Affiliation(s)
- Y Bakiri
- Department of Physiology, University College London, Gower Street, London, WC1E 6BT, UK
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
In making a selection of cellular tools and animal models for generating screening assays in the search for new drugs, one needs to take into consideration the practicality of their use in the drug discovery process. Conducting high-throughput primary screens using libraries of small molecules, close to 1 million members in size, requires the generation of large numbers of cells which are easily acquired, reliably enriched, and reproducibly responsive to standard positive controls. These cells need to be similar in form and function to their counterparts in human disease. In vitro assays that can be mechanized by using robots can therefore save time and costs. In selecting in vivo models, consideration must be given to the species and strain of animal chosen, the appropriateness of the model to human disease, the extent of animal husbandry required during the in-life pharmacological assessment, the technical aspects of generating the model and harvesting the tissues for analyses, the cost of research tools in terms of time and money (demyelinating and remyelinating agents, amount of compound to be generated), and the length of time required for drug testing in the model. A consideration of the translational aspects of the in vivo model compared to those used in the clinic is also important. These themes will be developed with examples for drug discovery in the field of CNS demyelination and repair, specifically as it pertains to multiple sclerosis.
Collapse
|
43
|
Lindahl JS, Kjellsen BR, Tigert J, Miskimins R. In utero PCP exposure alters oligodendrocyte differentiation and myelination in developing rat frontal cortex. Brain Res 2008; 1234:137-47. [PMID: 18675260 PMCID: PMC2572227 DOI: 10.1016/j.brainres.2008.06.126] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Revised: 06/25/2008] [Accepted: 06/27/2008] [Indexed: 01/02/2023]
Abstract
Several neurodevelopmental disorders, including schizophrenia, autism, ADD/ADHD and dyslexia are believed to originate during gestation and involve white matter abnormalities. Modulation of glutamate environments and glutamate receptors has also been implicated in alteration of oligodendrocytes, the myelin forming cells of the CNS. To begin to understand how modulation of the glutamate system affects the maturation of oligodendrocytes, developing rats were subjected to prenatal blockade of the NMDA receptor with phencyclidine (PCP). Oligodendrocyte development and differentiation were then examined postnatally by measuring markers for early, middle and late stage cells. The results indicate that, while the level of marker proteins for neurons and astrocytes remains the same, early oligodendrocyte progenitor cell markers are decreased in rat brains prenatally exposed to PCP. Labeling of cells of intermediate, immature cell stages is elevated. Late stage markers for myelinating oligodendrocytes are subsequently decreased. These data suggest that prenatal NMDA receptor blockade reduces the level of progenitors and that the surviving cells are arrested at an immature stage. This premature arrest appears to result in fewer fully differentiated, mature oligodendrocytes that are capable of producing myelin. These results have interesting implications for the role of glutamate and glutamate receptors in white matter abnormalities in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Josette S Lindahl
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, 414 E. Clark Street, Vermillion, SD 57069, USA
| | | | | | | |
Collapse
|
44
|
Delayed IGF-1 administration rescues oligodendrocyte progenitors from glutamate-induced cell death and hypoxic-ischemic brain damage. Dev Neurosci 2007; 29:302-10. [PMID: 17762198 DOI: 10.1159/000105471] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2007] [Accepted: 03/28/2007] [Indexed: 11/19/2022] Open
Abstract
We previously demonstrated that IGF-1 blocks glutamate-mediated death of late oligodendrocyte progenitors (OPs) by preventing Bax translocation, mitochondrial cytochrome c release and cleavage of caspases 9 and 3. Here, we demonstrate that IGF-1 prevents caspase 3 activation in late OPs when administered up to 16 h following exposure to glutamate. Moreover, late addition of IGF-1 to OPs previously exposed to toxic levels of glutamate promotes oligodendrocyte maturation as measured by myelin basic protein expression. We also demonstrate that intraventricularly administered IGF-1 retains OPs in the perinatal white matter after hypoxia-ischemia when given after insult. These results suggest that delayed administration of IGF-1 will rescue OPs in the immature white matter and promote myelination following hypoxia-ischemia.
Collapse
|
45
|
Matute C, Alberdi E, Domercq M, Sánchez-Gómez MV, Pérez-Samartín A, Rodríguez-Antigüedad A, Pérez-Cerdá F. Excitotoxic damage to white matter. J Anat 2007; 210:693-702. [PMID: 17504270 PMCID: PMC2375761 DOI: 10.1111/j.1469-7580.2007.00733.x] [Citation(s) in RCA: 194] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Glutamate kills neurons by excitotoxicity, which is caused by sustained activation of glutamate receptors. In recent years, it has been shown that glutamate can also be toxic to white matter oligodendrocytes and to myelin by this mechanism. In particular, glutamate receptor-mediated injury to these cells can be triggered by activation of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid, kainate and N-methyl-D-aspartate glutamate receptor types. Thus, these receptor classes, and the intermediaries of the signal cascades they activate, are potential targets for drug development to treat white matter damage in acute and chronic diseases. In addition, alterations of glutamate homeostasis in white matter can determine glutamate injury to oligodendrocytes and myelin. Astrocytes are responsible for most glutamate uptake in synaptic and non-synaptic areas and consequently are the major regulators of glutamate homeostasis. Activated microglia in turn may secrete cytokines and generate radical oxygen species, which impair glutamate uptake and reduce the expression of glutamate transporters. Finally, oligodendrocytes also contribute to glutamate homeostasis. This review aims at summarizing the current knowledge about the mechanisms leading to oligodendrocyte cell death and demyelination as a consequence of alterations in glutamate signalling, and their clinical relevance to disease. In addition, we show evidence that oligodendrocytes can also be killed by ATP acting at P2X receptors. A thorough understanding of how oligodendrocytes and myelin are damaged by excitotoxicity will generate knowledge that can lead to improved therapeutic strategies to protect white matter.
Collapse
Affiliation(s)
- Carlos Matute
- Departamento de Neurociencias, Universidad del País Vasco, Leioa, and Neurotek-UPV/EHU, Parque Tecnológico deBizkaia, Zamudio, Spain. carlos.,
| | | | | | | | | | | | | |
Collapse
|
46
|
McCarran WJ, Goldberg MP. White matter axon vulnerability to AMPA/kainate receptor-mediated ischemic injury is developmentally regulated. J Neurosci 2007; 27:4220-9. [PMID: 17429000 PMCID: PMC6672529 DOI: 10.1523/jneurosci.5542-06.2007] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Periventricular white matter injury (PWMI) is the leading cause of neurodevelopmental morbidity in survivors of premature birth. Cerebral ischemia is considered a major etiologic factor in the generation of PWMI. In adult white matter (WM), ischemic axonal damage is mediated by AMPA/kainate receptors. Mechanisms of ischemic axonal injury during development are not well defined. We used a murine brain slice model to characterize mechanisms of ischemic axonal injury in developing WM. Acute coronal brain slices were prepared from thy1-yellow fluorescent protein (YFP) mice at postnatal day 3 (P3), P7, P10, and P21. Ischemia was simulated by oxygen-glucose deprivation (OGD). YFP-positive axon morphology in the corpus callosum was preserved for at least 15 h under normoxic conditions. OGD resulted in delayed degeneration of YFP-positive axons, characterized by axonal beading, fragmentation, and loss of YFP. AMPA and cyclothiazide damaged WM axons at P7, P10, and P21 but not at P3. The AMPA/kainate receptor antagonist 2,3-dihydroxy-6-nitro-7-sulfamoyl-benzo[f]quinoxaline-2,3-dione (NBQX) decreased OGD-induced axonal degeneration and oligodendrocyte loss at P10 and P21. At P3 and P7, NBQX protected oligodendrocytes but did not prevent axonal degeneration after OGD. The NMDA receptor antagonist MK-801 [(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine maleate] provided no protection at any age. These results indicate that developing WM axons are susceptible to ischemic injury. However, mechanisms of axonal degeneration are developmentally regulated. At P3 and P7, corresponding developmentally to the window of peak vulnerability to PWMI in humans, ischemic axonal injury is not mediated by AMPA/kainate receptors. Strategies to protect WM during this period may be substantially different from those used at later developmental stages.
Collapse
Affiliation(s)
| | - Mark P. Goldberg
- Hope Center for Neurological Disorders and
- Neurology, Washington University School of Medicine, St. Louis, Missouri 63110
| |
Collapse
|
47
|
Abstract
Multiple sclerosis (MS) is a chronic demyelinating disease of the
human central nervous system (CNS). The condition predominantly
affects young adults and is characterised by immunological and
inflammatory changes in the periphery and CNS that contribute to
neurovascular disruption, haemopoietic cell invasion of target
tissues, and demyelination of nerve fibres which culminate in
neurological deficits that relapse and remit or are progressive.
The main features of MS can be reproduced in the inducible animal
counterpart, experimental autoimmune encephalomyelitis (EAE). The
search for new MS treatments invariably employs EAE to determine
drug activity and provide a rationale for exploring clinical
efficacy. The preclinical development of compounds for MS has
generally followed a conventional, immunotherapeutic route.
However, over the past decade, a group of compounds that suppress
EAE but have no apparent immunomodulatory activity have emerged.
These drugs interact with the N-methyl-D-aspartate (NMDA) and
α-amino-3-hydroxy-5-isoxazolepropionic acid (AMPA)/kainate
family of glutamate receptors reported to control neurovascular
permeability, inflammatory mediator synthesis, and resident glial
cell functions including CNS myelination. The review considers the
importance of the glutamate receptors in EAE and MS pathogenesis.
The use of receptor antagonists to control EAE is also discussed
together with the possibility of therapeutic application in
demyelinating disease.
Collapse
Affiliation(s)
- Christopher Bolton
- Centre for Biochemical Pharmacology and Experimental Pathology, John Vane Science
Centre, St Bartholomew's Hospital Medical School, Charterhouse Square, London EC1M 6BQ, UK
- *Christopher Bolton:
| | - Carolyn Paul
- Faculty of Applied Sciences, University of the West of England, Frenchay Campus,
Coldharbour Lane, Bristol BS16 1QY, UK
| |
Collapse
|
48
|
Redondo C, López-Toledano MA, Lobo MVT, Gonzalo-Gobernado R, Reimers D, Herranz AS, Paíno CL, Bazán E. Kainic acid triggers oligodendrocyte precursor cell proliferation and neuronal differentiation from striatal neural stem cells. J Neurosci Res 2007; 85:1170-82. [PMID: 17342781 DOI: 10.1002/jnr.21245] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Glutamate is an excitatory amino acid that serves important functions in mammalian brain development through alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA)/ kainate receptor stimulation. Neural stem cells with self-renewal and multilineage potential are a useful tool to study the signals involved in the regulation of brain development. We have investigated the role played by AMPA/kainate receptors during the differentiation of neural stem cells derived from fetal rat striatum. The application of 1 and 10 microM kainic acid increased significantly the phosphorylation of the cyclic AMP response element binding protein (CREB), raised bromodeoxyuridine incorporation in O4-positive oligodendrocyte precursors, and increased the number of O1-positive cells in the cultures. Increased CREB phosphorylation and proliferation were prevented by the AMPA receptor antagonist 4-4(4-aminophenyl)-1,2-dihydro-1-methyl-2-propylcarbamoyl-6,7-methylenedioxyphthalazine (SYM 2206) and by protein kinase A and protein kinase C inhibitors. Cultures treated with 100 microM kainic acid showed decreased proliferation, a lower proportion of O1-positive cells, and apoptosis of O4-positive cells. None of these effects were prevented by SYM 2206, suggesting that kainate receptors take part in these events. We conclude that AMPA receptor stimulation by kainic acid promotes the proliferation of oligodendrocyte precursors derived from neural stem cells through a mechanism that requires the activation of CREB by protein kinase A and C. In the neurons derived from these cells, either AMPA or kainate receptor stimulation produces neuritic growth and larger cell bodies.
Collapse
Affiliation(s)
- Carolina Redondo
- Servicio de Neurobiología, Departamento de Investigación, Hospital Ramón y Cajal, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Benjamins JA, Nedelkoska L. Cyclic GMP-dependent pathways protect differentiated oligodendrocytes from multiple types of injury. Neurochem Res 2006; 32:321-9. [PMID: 17191140 DOI: 10.1007/s11064-006-9187-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2006] [Accepted: 09/27/2006] [Indexed: 12/13/2022]
Abstract
The cyclic GMP analog 8-bromo-cyclic GMP (8-Br-cGMP) protects differentiated murine oligodendrocytes (OLs) from caspase-mediated death initiated by staurosporine, thapsigargin or kainate. Caspase-independent death caused by high levels of NO is also partially prevented by 8-Br-cGMP. Inhibitors of protein kinase G (cGMP-dependent protein kinase, cGK) reversed protection, supporting involvement of cGK. Since NO stimulates soluble guanylate cyclase, increasing cGMP, we treated OLs with low levels of NO and observed partial protection against thapsigargin, staurosporine and kainate. Two inhibitors of mitochondrial pore transition (MPT), cyclosporin A and bongkrekic acid, were poorly protective, indicating that cGMP is not acting primarily by blocking MPT. 8Br-cGMP was more effective than 8Br-cAMP in protecting against staurosporine or release of intracellular Ca(++) by thapsigargin. The cAMP analog exhibited little or no protection against kainate or high levels of NO. Thus cGK signaling is more effective than protein kinase A or phosphodiesterase 3 signaling in preventing OL death.
Collapse
Affiliation(s)
- Joyce A Benjamins
- Department of Neurology, Wayne State University School of Medicine, 1228 Elliman Building, 421 E. Canfield Avenue, Detroit, MI 48201, USA.
| | | |
Collapse
|
50
|
Underhill SM, Goldberg MP. Hypoxic injury of isolated axons is independent of ionotropic glutamate receptors. Neurobiol Dis 2006; 25:284-90. [PMID: 17071096 PMCID: PMC1892630 DOI: 10.1016/j.nbd.2006.09.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2006] [Revised: 09/05/2006] [Accepted: 09/16/2006] [Indexed: 10/24/2022] Open
Abstract
Axonal injury in white matter is an important consequence of many acute neurological diseases including ischemia. A role for glutamate-mediated excitotoxicity is suggested by observations from in vitro and in situ models that AMPA/kainate blockers can reduce axonal injury. We assessed axonal vulnerability in primary murine neuronal cultures, with axons isolated from their cell bodies using a compartmented chamber design. Transient removal of oxygen and glucose in the axon compartment resulted in irreversible loss of axon length and neurofilament labeling. This injury was not prevented by addition of ionotropic glutamate receptor blockers and could not be reproduced by glutamate receptor agonists. However, hypoxic injury was prevented by blockade of voltage-gated sodium channels, inhibition of calpain and removal of extracellular calcium. These results suggest that isolated, unmyelinated axons are vulnerable to hypoxic injury which is mediated by influx of sodium and calcium but is independent of glutamate receptor activation.
Collapse
Affiliation(s)
- Suzanne M Underhill
- Hope Center for Neurological Disorders and Department of Neurology, 660 S. Euclid Avenue, Campus Box 8111, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | |
Collapse
|