1
|
Musotto R, Wanderlingh U, Pioggia G. Ca 2+ waves in astrocytes: computational modeling and experimental data. Front Cell Neurosci 2025; 19:1536096. [PMID: 40226297 PMCID: PMC11985530 DOI: 10.3389/fncel.2025.1536096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/06/2025] [Indexed: 04/15/2025] Open
Abstract
This paper examines different computational models for Calcium wave propagation in astrocytes. Through a comparative analysis of models by Goldbeter, De Young-Keizer, Atri, Li-Rinzel, and De Pittà and of experimental data, the study highlights the model contributions for the understanding of Calcium dynamics. Tracing the evolution from simple to complex models, this work emphasizes the importance of integrating experimental data in order to further refine these models. The results allow to improve our understanding of the physiological functions of astrocytes, suggesting the importance of more accurate astrocyte models.
Collapse
Affiliation(s)
- Rosa Musotto
- National Research Council, IRIB-CNR, Institute for Biomedical Research and Innovation, Messina, Italy
| | - Ulderico Wanderlingh
- Department of Mathematical and Computer Sciences, Physical Sciences and Earth Sciences, University of Messina, Messina, Italy
| | - Giovanni Pioggia
- National Research Council, IRIB-CNR, Institute for Biomedical Research and Innovation, Messina, Italy
| |
Collapse
|
2
|
Liu X, Ying J, Wang X, Zheng Q, Zhao T, Yoon S, Yu W, Yang D, Fang Y, Hua F. Astrocytes in Neural Circuits: Key Factors in Synaptic Regulation and Potential Targets for Neurodevelopmental Disorders. Front Mol Neurosci 2021; 14:729273. [PMID: 34658786 PMCID: PMC8515196 DOI: 10.3389/fnmol.2021.729273] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/02/2021] [Indexed: 12/14/2022] Open
Abstract
Astrocytes are the major glial cells in the brain, which play a supporting role in the energy and nutritional supply of neurons. They were initially regarded as passive space-filling cells, but the latest progress in the study of the development and function of astrocytes highlights their active roles in regulating synaptic transmission, formation, and plasticity. In the concept of "tripartite synapse," the bidirectional influence between astrocytes and neurons, in addition to their steady-state and supporting function, suggests that any negative changes in the structure or function of astrocytes will affect the activity of neurons, leading to neurodevelopmental disorders. The role of astrocytes in the pathophysiology of various neurological and psychiatric disorders caused by synaptic defects is increasingly appreciated. Understanding the roles of astrocytes in regulating synaptic development and the plasticity of neural circuits could help provide new treatments for these diseases.
Collapse
Affiliation(s)
- Xing Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Jun Ying
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Xifeng Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qingcui Zheng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Tiancheng Zhao
- Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Sungtae Yoon
- Helping Minds International Charitable Foundation, New York, NY, United States
| | - Wen Yu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Danying Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Yang Fang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| |
Collapse
|
3
|
Yeung JHY, Walby JL, Palpagama TH, Turner C, Waldvogel HJ, Faull RLM, Kwakowsky A. Glutamatergic receptor expression changes in the Alzheimer's disease hippocampus and entorhinal cortex. Brain Pathol 2021; 31:e13005. [PMID: 34269494 PMCID: PMC8549033 DOI: 10.1111/bpa.13005] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/25/2021] [Accepted: 06/21/2021] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's Disease (AD) is the leading form of dementia worldwide. Currently, the pathological mechanisms underlying AD are not well understood. Although the glutamatergic system is extensively implicated in its pathophysiology, there is a gap in knowledge regarding the expression of glutamate receptors in the AD brain. This study aimed to characterize the expression of specific glutamate receptor subunits in post‐mortem human brain tissue using immunohistochemistry and confocal microscopy. Free‐floating immunohistochemistry and confocal laser scanning microscopy were used to quantify the density of glutamate receptor subunits GluA2, GluN1, and GluN2A in specific cell layers of the hippocampal sub‐regions, subiculum, entorhinal cortex, and superior temporal gyrus. Quantification of GluA2 expression in human post‐mortem hippocampus revealed a significant increase in the stratum (str.) moleculare of the dentate gyrus (DG) in AD compared with control. Increased GluN1 receptor expression was found in the str. moleculare and hilus of the DG, str. oriens of the CA2 and CA3, str. pyramidale of the CA2, and str. radiatum of the CA1, CA2, and CA3 subregions and the entorhinal cortex. GluN2A expression was significantly increased in AD compared with control in the str. oriens, str. pyramidale, and str. radiatum of the CA1 subregion. These findings indicate that the expression of glutamatergic receptor subunits shows brain region‐specific changes in AD, suggesting possible pathological receptor functioning. These results provide evidence of specific glutamatergic receptor subunit changes in the AD hippocampus and entorhinal cortex, indicating the requirement for further research to elucidate the pathophysiological mechanisms it entails, and further highlight the potential of glutamatergic receptor subunits as therapeutic targets.
Collapse
Affiliation(s)
- Jason H Y Yeung
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Joshua L Walby
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Thulani H Palpagama
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Clinton Turner
- Department of Anatomical Pathology, LabPlus, Auckland City Hospital, Auckland, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
4
|
Yeung JHY, Calvo-Flores Guzmán B, Palpagama TH, Ethiraj J, Zhai Y, Tate WP, Peppercorn K, Waldvogel HJ, Faull RLM, Kwakowsky A. Amyloid-beta 1-42 induced glutamatergic receptor and transporter expression changes in the mouse hippocampus. J Neurochem 2020; 155:62-80. [PMID: 32491248 DOI: 10.1111/jnc.15099] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is the leading type of dementia worldwide. With an increasing burden of an aging population coupled with the lack of any foreseeable cure, AD warrants the current intense research effort on the toxic effects of an increased concentration of beta-amyloid (Aβ) in the brain. Glutamate is the main excitatory brain neurotransmitter and it plays an essential role in the function and health of neurons and neuronal excitability. While previous studies have shown alterations in expression of glutamatergic signaling components in AD, the underlying mechanisms of these changes are not well understood. This is the first comprehensive anatomical study to characterize the subregion- and cell layer-specific long-term effect of Aβ1-42 on the expression of specific glutamate receptors and transporters in the mouse hippocampus, using immunohistochemistry with confocal microscopy. Outcomes are examined 30 days after Aβ1-42 stereotactic injection in aged male C57BL/6 mice. We report significant decreases in density of the glutamate receptor subunit GluA1 and the vesicular glutamate transporter (VGluT) 1 in the conus ammonis 1 region of the hippocampus in the Aβ1-42 injected mice compared with artificial cerebrospinal fluid injected and naïve controls, notably in the stratum oriens and stratum radiatum. GluA1 subunit density also decreased within the dentate gyrus dorsal stratum moleculare in Aβ1-42 injected mice compared with artificial cerebrospinal fluid injected controls. These changes are consistent with findings previously reported in the human AD hippocampus. By contrast, glutamate receptor subunits GluA2, GluN1, GluN2A, and VGluT2 showed no changes in expression. These findings indicate that Aβ1-42 induces brain region and layer specific expression changes of the glutamatergic receptors and transporters, suggesting complex and spatial vulnerability of this pathway during development of AD neuropathology. Read the Editorial Highlight for this article on page 7. Cover Image for this issue: https://doi.org/10.1111/jnc.14763.
Collapse
Affiliation(s)
- Jason H Y Yeung
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Beatriz Calvo-Flores Guzmán
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Thulani H Palpagama
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Jayarjun Ethiraj
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Ying Zhai
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Warren P Tate
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Katie Peppercorn
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
5
|
Quercetin mitigates monosodium glutamate-induced excitotoxicity of the spinal cord motoneurons in aged rats via p38 MAPK inhibition. Acta Histochem 2020; 122:151554. [PMID: 32622428 DOI: 10.1016/j.acthis.2020.151554] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/19/2020] [Accepted: 04/22/2020] [Indexed: 12/18/2022]
Abstract
Various studies reported the possibility of deterioration of blood-brain barrier (BBB) integrity owing to the aging process. The current work was performed to investigate the ability of Monosodium glutamate (MSG) to cross BBB in aged rats, the damage affecting the anterior horn cells of the spinal cord due to excitotoxicity, and the mechanisms by which quercetin (Que) administration might suppress such damage. Forty male rats aged 18 months were assigned equally to 4 groups: control group, Que group (received Que, 20 mg/kg/d intraperitonealy for 10 days), MSG group (received MSG, 4.0 g/kg/d subcutaneously for 10 days), MSG + Que group (received both Que and MSG as done in the Que and MSG groups respectively). Cervical spinal cord specimens were obtained and prepared for routine histological study, immunohistochemical staining by caspase-3 and glial fibrillary acidic protein (GFAP), assessment of oxidative stress, measurement of cytokines, assessment of caspase-3 activity and GFAP levels as well as for western blotting of phosphorylated activating transcription factor 2 (ATF2pp) as an indicator for the activity of p38 mitogen-activated protein kinase (MAPK). The MSG group revealed variable degenerative and apoptotic changes in the motoneurons and neuroglia, a marked rise in the cytoplasmic caspase-3 expression in motoneurons and a significant reduction (p < 0.001) in the astrocyte surface area percentage. In addition, the spinal cord tissue exhibited a significant elevation (p < 0.001) in the levels of malondialdehyde (MDA), IL-1, IL-6, TNFα, INFɣ, caspase-3 activity and ATF2 pp expression as well as a significant reduction (p < 0.001) in SOD, IL-10 and GFAP levels compared with the control group. On combining Que with MSG, most of the degenerative changes were reversed and all the impaired parameters were nearly normalized except for IL-6 and GFAP levels which were still significantly (p < 0.05) different from those of the control group. Our study suggests that MSG can break through the BBB of the aged rats and induce excitotoxicity dependent changes in spinal cord motoneurons. Most of these changes were reversed by Que probably via targeting the p38 MAPK-ATF2 pathway, antagonizing oxidative stress, anti-inflammatory effect, and promoting GFAP expression.
Collapse
|
6
|
Beiersdorfer A, Lohr C. AMPA Receptor-Mediated Ca 2+ Transients in Mouse Olfactory Ensheathing Cells. Front Cell Neurosci 2019; 13:451. [PMID: 31636544 PMCID: PMC6788192 DOI: 10.3389/fncel.2019.00451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/20/2019] [Indexed: 11/13/2022] Open
Abstract
Ca2+ signaling in glial cells is primarily triggered by metabotropic pathways and the subsequent Ca2+ release from internal Ca2+ stores. However, there is upcoming evidence that various ion channels might also initiate Ca2+ rises in glial cells by Ca2+ influx. We investigated AMPA receptor-mediated inward currents and Ca2+ transients in olfactory ensheathing cells (OECs), a specialized glial cell population in the olfactory bulb (OB), using whole-cell voltage-clamp recordings and confocal Ca2+ imaging. By immunohistochemistry we showed immunoreactivity to the AMPA receptor subunits GluA1, GluA2 and GluA4 in OECs, suggesting the presence of AMPA receptors in OECs. Kainate-induced inward currents were mediated exclusively by AMPA receptors, as they were sensitive to the specific AMPA receptor antagonist, GYKI53655. Moreover, kainate-induced inward currents were reduced by the selective Ca2+-permeable AMPA receptor inhibitor, NASPM, suggesting the presence of functional Ca2+-permeable AMPA receptors in OECs. Additionally, kainate application evoked Ca2+ transients in OECs which were abolished in the absence of extracellular Ca2+, indicating that Ca2+ influx via Ca2+-permeable AMPA receptors contribute to kainate-induced Ca2+ transients. However, kainate-induced Ca2+ transients were partly reduced upon Ca2+ store depletion, leading to the conclusion that Ca2+ influx via AMPA receptor channels is essential to trigger Ca2+ transients in OECs, whereas Ca2+ release from internal stores contributes in part to the kainate-evoked Ca2+ response. Endogenous glutamate release by OSN axons initiated Ca2+ transients in OECs, equally mediated by metabotropic receptors (glutamatergic and purinergic) and AMPA receptors, suggesting a prominent role for AMPA receptor mediated Ca2+ signaling in axon-OEC communication.
Collapse
Affiliation(s)
| | - Christian Lohr
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| |
Collapse
|
7
|
Kumar M, John M, Madhavan M, James J, Omkumar RV. Alteration in the phosphorylation status of NMDA receptor GluN2B subunit by activation of both NMDA receptor and L-type voltage gated calcium channel. Neurosci Lett 2019; 709:134343. [PMID: 31279915 DOI: 10.1016/j.neulet.2019.134343] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/03/2019] [Accepted: 06/18/2019] [Indexed: 01/27/2023]
Abstract
Calcium influx through N-methyl-D-aspartate receptors (NMDAR) and voltage-gated calcium channels (VGCC) play major roles in postsynaptic signaling mechanisms. NMDAR subunit GluN2B is phosphorylated at Ser1303. Phosphorylation at this site is a prominent event in cell culture systems as well as in vivo. However, the functional significance of phosphorylation at this site is not completely understood. In this study, we compared the effect of calcium signaling through NMDAR and VGCC on the phosphorylation status of GluN2B-Ser1303 in the rat in vivo. VGCC was activated by intraperitoneal (IP) injection of the activator, BayK8644 and NMDAR was activated by intracerebroventricular (ICV) injection of NMDA in separate experimental groups. We found that the level of phospho-GluN2B-Ser1303 in the cortex and in the hippocampus increased in response to activation of either channel. The effects could be prevented by prior ICV administration of the specific blockers of these channels such as MK-801 for NMDAR and nifedipine for VGCC. The effect was also blocked by pretreatment with ICV administration of KN-93 indicating that it is mediated through CaM kinase. Both during NMDAR activation and VGCC activation, cell survival associated signals such as phospho-AKT and phospho-CREB showed decrease, consistent with activation of cell death pathways during these treatments. We conclude that under in vivo conditions, calcium influx through either NMDAR or VGCC activates CaM kinase, which in turn phosphorylates GluN2B-Ser1303.
Collapse
Affiliation(s)
- Mantosh Kumar
- Molecular Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thycaud, P.O., Thiruvananthapuram-695014, Kerala, India; Research Scholar, University of Kerala, India
| | - Mathew John
- Molecular Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thycaud, P.O., Thiruvananthapuram-695014, Kerala, India
| | - Mayadevi Madhavan
- Molecular Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thycaud, P.O., Thiruvananthapuram-695014, Kerala, India
| | - Jackson James
- Neuro Stem Cell Biology Lab, Rajiv Gandhi Centre for Biotechnology, India
| | - Ramakrishnapillai V Omkumar
- Molecular Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thycaud, P.O., Thiruvananthapuram-695014, Kerala, India.
| |
Collapse
|
8
|
Farag A, Lashen S, Eltaysh R. Histoarchitecture restoration of cerebellar sub-layers as a response to estradiol treatment following Kainic acid-induced spinal cord injury. Cell Tissue Res 2019; 376:309-323. [PMID: 30788578 DOI: 10.1007/s00441-019-02992-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 12/29/2018] [Indexed: 12/19/2022]
Abstract
One of the major impacts of spinal cord injury (SCI) is the cerebellar neurological malfunction and deformation of its sub-layers. This could be due to the enormous innervation of the spinocerebellar tract from the posterior gray horn in the spinal cord to the ipsilateral cerebellum. Although the neuroprotective role of estradiol in spinal cord (SC) injuries, as well as its ability to delay secondary cell death changes, is well-known, its effect on cerebellar layers is not fully investigated. In this study, a SCI model was achieved by injection of Kainic acid into SC of adult Male Wistar rats in order to assess the effects of SCI on the cerebellum. The animals were classified into SCI group (animals with SCI), estradiol-treated group (animals with SCI and received estradiol), control groups, and sham control group. The microscopical examination 24 h after induction of SCI revealed that KA induced the most characteristics of neurodegeneration including astrocytic propagation and microglial activation. The estradiol was injected intraperitoneally 20 min after induction of SCI, and the samples were collected at 1, 3, 7, 14, and 30 days. Histologically, the estradiol reduced the inflammatory response, enhanced the recovery of molecular, granular, and Purkinje cell layers, and therefore aided in the restoration of layer organization. These findings were also confirmed by immunohistochemical staining and gene expression profiling.
Collapse
Affiliation(s)
- Amany Farag
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Gomhoria St., P.O. box 35516, Mansoura, Egypt.
| | - S Lashen
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Gomhoria St., P.O. box 35516, Mansoura, Egypt
| | - R Eltaysh
- Department of Pharmacology, Faculty of Veterinary Medicine, Mansoura University, Gomhoria St., P.O. box 35516, Mansoura, Egypt
| |
Collapse
|
9
|
Herrera-Zamora JM, Castro-Sánchez LA, Reyes-Mendez M, Aguilar-Martinez I, Osuna-López F, Moreno-Galindo EG, Navarro-Polanco RA, Aguilar-Roblero RA, Sánchez-Pastor E, Alamilla J. Development-Dependent Changes in the NR2 Subtype of the N-Methyl-D-Aspartate Receptor in the Suprachiasmatic Nucleus of the Rat. J Biol Rhythms 2019; 34:39-50. [DOI: 10.1177/0748730418824198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- J. Manuel Herrera-Zamora
- Centro Universitario de Investigaciones Biomédicas “CUIB”, Universidad de Colima, Colima, Col, Mexico
| | - Luis A. Castro-Sánchez
- Centro Universitario de Investigaciones Biomédicas “CUIB”, Universidad de Colima, Colima, Col, Mexico
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Universidad de Colima, Colima, Col, Mexico
| | - Miriam Reyes-Mendez
- Centro Universitario de Investigaciones Biomédicas “CUIB”, Universidad de Colima, Colima, Col, Mexico
| | - Irving Aguilar-Martinez
- Centro Universitario de Investigaciones Biomédicas “CUIB”, Universidad de Colima, Colima, Col, Mexico
| | - Fernando Osuna-López
- Centro Universitario de Investigaciones Biomédicas “CUIB”, Universidad de Colima, Colima, Col, Mexico
| | - Eloy G. Moreno-Galindo
- Centro Universitario de Investigaciones Biomédicas “CUIB”, Universidad de Colima, Colima, Col, Mexico
| | | | - Raul A. Aguilar-Roblero
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México
| | - Enrique Sánchez-Pastor
- Centro Universitario de Investigaciones Biomédicas “CUIB”, Universidad de Colima, Colima, Col, Mexico
| | - Javier Alamilla
- Centro Universitario de Investigaciones Biomédicas “CUIB”, Universidad de Colima, Colima, Col, Mexico
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Universidad de Colima, Colima, Col, Mexico
| |
Collapse
|
10
|
Harnisch K, Teuber-Hanselmann S, Macha N, Mairinger F, Fritsche L, Soub D, Meinl E, Junker A. Myelination in Multiple Sclerosis Lesions Is Associated with Regulation of Bone Morphogenetic Protein 4 and Its Antagonist Noggin. Int J Mol Sci 2019; 20:ijms20010154. [PMID: 30609838 PMCID: PMC6337410 DOI: 10.3390/ijms20010154] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 12/22/2018] [Accepted: 12/27/2018] [Indexed: 11/25/2022] Open
Abstract
Remyelination is a central aspect of new multiple sclerosis (MS) therapies, in which one aims to alleviate disease symptoms by improving axonal protection. However, a central problem is mediators expressed in MS lesions that prevent effective remyelination. Bone morphogenetic protein4 (BMP4) inhibits the development of mature oligodendrocytes in cell culture and also blocks the expression of myelin proteins. Additionally, numerous studies have shown that Noggin (SYM1)—among other physiological antagonists of BMP4—plays a prominent role in myelin formation in the developing but also the adult central nervous system. Nonetheless, neither BMP4 nor Noggin have been systematically studied in human MS lesions. In this study, we demonstrated by transcript analysis and immunohistochemistry that BMP4 is expressed by astrocytes and microglia/macrophages in association with inflammatory infiltrates in MS lesions, and that astrocytes also express BMP4 in chronic inactive lesions that failed to remyelinate. Furthermore, the demonstration of an increased expression of Noggin in so-called shadow plaques (i.e., remyelinated lesions with thinner myelin sheaths) in comparison to chronically inactive demyelinated lesions implies that antagonizing BMP4 is associated with successful remyelination in MS plaques in humans. However, although BMP4 is strongly overexpressed in inflammatory lesion areas, its levels are also elevated in remyelinated lesion areas, which raises the possibility that BMP4 signaling itself may be required for remyelination. Therefore, remyelination might be influenced by a small number of key factors. Manipulating these molecules, i.e., BMP4 and Noggin, could be a promising therapeutic approach for effective remyelination.
Collapse
Affiliation(s)
- Kim Harnisch
- Institute of Neuropathology, University Hospital Essen, D-45147 Essen, Germany.
| | | | - Nicole Macha
- Institute of Neuropathology, University Hospital Essen, D-45147 Essen, Germany.
| | - Fabian Mairinger
- Institute of Pathology, University Hospital Essen, D-45147 Essen, Germany.
| | - Lena Fritsche
- Institute of Neuropathology, University Hospital Essen, D-45147 Essen, Germany.
| | - Daniel Soub
- Institute of Neuropathology, University Hospital Essen, D-45147 Essen, Germany.
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians-Universität München, D-82152 Martinsried, Germany.
| | - Andreas Junker
- Institute of Neuropathology, University Hospital Essen, D-45147 Essen, Germany.
| |
Collapse
|
11
|
Colombo JA. Cellular complexity in subcortical white matter: a distributed control circuit? Brain Struct Funct 2018; 223:981-985. [DOI: 10.1007/s00429-018-1609-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 01/10/2018] [Indexed: 11/30/2022]
|
12
|
Berret E, Barron T, Xu J, Debner E, Kim EJ, Kim JH. Oligodendroglial excitability mediated by glutamatergic inputs and Nav1.2 activation. Nat Commun 2017; 8:557. [PMID: 28916793 PMCID: PMC5601459 DOI: 10.1038/s41467-017-00688-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 07/19/2017] [Indexed: 12/19/2022] Open
Abstract
Oligodendrocyte (OL) maturation and axon-glial communication are required for proper myelination in the developing brain. However, physiological properties of OLs remain largely uncharacterized in different brain regions. The roles of oligodendroglial voltage-activated Na+ channels (Nav) and electrical excitability in relation to maturation to the myelinating stage are controversial, although oligodendroglial excitability is potentially important for promoting axon myelination. Here we show spiking properties of OLs and their role in axon-glial communication in the auditory brainstem. A subpopulation of pre-myelinating OLs (pre-OLs) can generate Nav1.2-driven action potentials throughout postnatal development to early adulthood. In addition, excitable pre-OLs receive glutamatergic inputs from neighboring neurons that trigger pre-OL spikes. Knockdown of Nav1.2 channels in pre-OLs alters their morphology, reduces axon-OL interactions and impairs myelination. Our results suggest that Nav1.2-driven spiking of pre-OLs is an integral component of axon-glial communication and is required for the function and maturation of OLs to promote myelination.Axon-glial communication is important for myelination. Here the authors show that during postnatal development in rats, a subpopulation of pre-myelinating oligodendrocytes in the auditory brainstem receive excitatory inputs and can generate Nav 1.2-driven action potentials, and that such process promotes myelination.
Collapse
Affiliation(s)
- Emmanuelle Berret
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center, San Antonio, Texas, 78229, USA
| | - Tara Barron
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center, San Antonio, Texas, 78229, USA
| | - Jie Xu
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center, San Antonio, Texas, 78229, USA
| | - Emily Debner
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center, San Antonio, Texas, 78229, USA
| | - Eun Jung Kim
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center, San Antonio, Texas, 78229, USA
| | - Jun Hee Kim
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center, San Antonio, Texas, 78229, USA.
| |
Collapse
|
13
|
Toma VA, Farcas AD, Parvu M, Silaghi-Dumitrescu R, Roman I. CA3 hippocampal field: Cellular changes and its relation with blood nitro-oxidative stress reveal a balancing function of CA3 area in rats exposed to repetead restraint stress. Brain Res Bull 2016; 130:10-17. [PMID: 28013041 DOI: 10.1016/j.brainresbull.2016.12.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 10/29/2016] [Accepted: 12/20/2016] [Indexed: 12/23/2022]
Abstract
Rats exposed to repeated restraint stress exhibit structural and functional deficits in hippocampus that are similar to those observed in patients with depressive illnesses. Blood corticosterone concentrations are proportionally increased with catalase and glutathione-peroxidase activity and are inversely proportional with 3-nitrotyrosine concentrations.Cytochrome c oxidase, adenosin tryphosphatase and monoamine oxidase activities of CA3 hippocampal field mark a stress-time dependent decrease. Acridine-orange labeling of the CA3 field reveals an enhancing green fluorescence of glyocites in stress conditions. After three days of restraint stress, the secretory activity of CA3 neurons did not show significant decrease, and neurons appeared with normal shapes and distribution. CA3 neurons after seven days of restraint stress have marked a slight decrease of secretory activity. In contrast to a well-preserved histological appearance of the CA3 neurons, local and blood stress-related reactions are observed. CA3-glial activation and disturbance of blood oxidative homeostasis are tandem processes during three and seven days of RS. This study depicts the balancing role of CA3 area in time-varying stress conditions.
Collapse
Affiliation(s)
- Vlad Al Toma
- Babes-Bolyai University, Cluj-Napoca, Romania; Institute of Biological Research, Cluj-Napoca, Romania; NIRD of Isotopic and Molecular Technologies, Cluj-Napoca, Romania.
| | - Anca D Farcas
- Babes-Bolyai University, Cluj-Napoca, Romania; Institute of Biological Research, Cluj-Napoca, Romania; NIRD of Isotopic and Molecular Technologies, Cluj-Napoca, Romania
| | | | | | - Ioana Roman
- Institute of Biological Research, Cluj-Napoca, Romania
| |
Collapse
|
14
|
Glia plasma membrane transporters: Key players in glutamatergic neurotransmission. Neurochem Int 2016; 98:46-55. [DOI: 10.1016/j.neuint.2016.04.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 03/07/2016] [Accepted: 04/06/2016] [Indexed: 12/27/2022]
|
15
|
Abstract
The amino acid L-Glutamate acts as the most ubiquitous mediator of excitatory synaptic transmission in the central nervous system. Glutamatergic transmission is central for diverse brain functions, being particularly important for learning, memory, and cognition. In brain pathology, excessive release of glutamate triggers excitotoxic neural cell death through necrotic or apoptotic pathways. Glutamate effects are mediated by several classes of glutamate receptors, expressed in virtually all cells of neural origin. Specifically important for both physiological information processing and cell damage are glutamate receptors of NMDA ( N-methyl-D-aspartate) type, which, for a long time, were considered to be expressed exclusively in neurons. Recent studies have found functional NMDA receptors in brain macroglia, in astrocytes, and oligodendrocytes. Glial and neuronal NMDA receptors are functionally and structurally different; the glial receptors are weakly (if at all) sensitive to the extracellular magnesium block, which may indicate a predominant expression of the NR3 receptor subunit. In the cortex, astroglial NMDA receptors are activated upon physiological synaptic transmission. The physiological relevance of NMDA receptors in the white matter remains unknown; their activation upon ischemia triggers Ca2+-dependent damage of oligodendrocytes and myelin. The discovery of glial NMDA receptors further indicates the complex nature of intercellular signaling mechanisms in the brain, which involve all types of neural cells, connected through diverse types of chemical and electrical synapses.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, 1.124 Stopford Building, Oxford Road, Manchester M13 9PT UK.
| | | |
Collapse
|
16
|
Glutamate and ATP at the Interface Between Signaling and Metabolism in Astroglia: Examples from Pathology. Neurochem Res 2016; 42:19-34. [PMID: 26915104 DOI: 10.1007/s11064-016-1848-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 01/21/2016] [Accepted: 01/22/2016] [Indexed: 12/17/2022]
Abstract
Glutamate is the main excitatory transmitter in the brain, while ATP represents the most important energy currency in any living cell. Yet, these chemicals play an important role in both processes, enabling them with dual-acting functions in metabolic and intercellular signaling pathways. Glutamate can fuel ATP production, while ATP can act as a transmitter in intercellular signaling. We discuss the interface between glutamate and ATP in signaling and metabolism of astrocytes. Not only do glutamate and ATP cross each other's paths in physiology of the brain, but they also do so in its pathology. We present the fabric of this process in (patho)physiology through the discussion of synthesis and metabolism of ATP and glutamate in astrocytes as well as by providing a general description of astroglial receptors for these molecules along with the downstream signaling pathways that may be activated. It is astroglial receptors for these dual-acting molecules that could hold a key for medical intervention in pathological conditions. We focus on two examples disclosing the role of activation of astroglial ATP and glutamate receptors in pathology of two kinds of brain tissue, gray matter and white matter, respectively. Interventions at the interface of metabolism and signaling show promise for translational medicine.
Collapse
|
17
|
Zhou Y, Danbolt NC. Glutamate as a neurotransmitter in the healthy brain. J Neural Transm (Vienna) 2014; 121:799-817. [PMID: 24578174 PMCID: PMC4133642 DOI: 10.1007/s00702-014-1180-8] [Citation(s) in RCA: 585] [Impact Index Per Article: 53.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 02/11/2014] [Indexed: 12/13/2022]
Abstract
Glutamate is the most abundant free amino acid in the brain and is at the crossroad between multiple metabolic pathways. Considering this, it was a surprise to discover that glutamate has excitatory effects on nerve cells, and that it can excite cells to their death in a process now referred to as "excitotoxicity". This effect is due to glutamate receptors present on the surface of brain cells. Powerful uptake systems (glutamate transporters) prevent excessive activation of these receptors by continuously removing glutamate from the extracellular fluid in the brain. Further, the blood-brain barrier shields the brain from glutamate in the blood. The highest concentrations of glutamate are found in synaptic vesicles in nerve terminals from where it can be released by exocytosis. In fact, glutamate is the major excitatory neurotransmitter in the mammalian central nervous system. It took, however, a long time to realize that. The present review provides a brief historical description, gives a short overview of glutamate as a transmitter in the healthy brain, and comments on the so-called glutamate-glutamine cycle. The glutamate transporters responsible for the glutamate removal are described in some detail.
Collapse
Affiliation(s)
- Y. Zhou
- The Neurotransporter Group, Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, Blindern, P.O. Box 1105, 0317 Oslo, Norway
| | - N. C. Danbolt
- The Neurotransporter Group, Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, Blindern, P.O. Box 1105, 0317 Oslo, Norway
| |
Collapse
|
18
|
The role of astrocytes in the regulation of synaptic plasticity and memory formation. Neural Plast 2013; 2013:185463. [PMID: 24369508 PMCID: PMC3867861 DOI: 10.1155/2013/185463] [Citation(s) in RCA: 133] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 10/07/2013] [Accepted: 11/05/2013] [Indexed: 12/22/2022] Open
Abstract
Astrocytes regulate synaptic transmission and play a role in the formation of new memories, long-term potentiation (LTP), and functional synaptic plasticity. Specifically, astroglial release of glutamate, ATP, and cytokines likely alters the survivability and functioning of newly formed connections. Among these pathways, regulation of glutamate appears to be most directly related to the promotion of LTP, which is highly dependent on the synchronization of synaptic receptors through the regulation of excitatory postsynaptic potentials. Moreover, regulation of postsynaptic glutamate receptors, particularly AMPA receptors, is dependent on signaling by ATP synthesized in astrocytes. Finally, cytokine signaling is also implicated in regulating LTP, but is likely most important in plasticity following tissue damage. Despite the role of these signaling factors in regulating LTP and functional plasticity, an integrative model of these factors has not yet been elucidated. In this review, we seek to summarize the current body of evidence on astrocytic mechanisms for regulation of LTP and functional plasticity, and provide an integrative model of the processes.
Collapse
|
19
|
Glutamate transporters in the biology of malignant gliomas. Cell Mol Life Sci 2013; 71:1839-54. [PMID: 24281762 DOI: 10.1007/s00018-013-1521-z] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 10/22/2013] [Accepted: 11/11/2013] [Indexed: 12/11/2022]
Abstract
Malignant gliomas are relentless tumors that offer a dismal clinical prognosis. They develop many biological advantages that allow them to grow and survive in the unique environment of the brain. The glutamate transporters system x c (-) and excitatory amino acid transporters (EAAT) are emerging as key players in the biology and malignancy of these tumors. Gliomas manipulate glutamate transporter expression and function to alter glutamate homeostasis in the brain, which supports their own growth, invasion, and survival. As a consequence, malignant cells are able to quickly destroy and invade surrounding normal brain. Recent findings are painting a larger picture of these transporters in glioma biology, and as such are providing opportunities for clinical intervention for patients. This review will detail the current understanding of glutamate transporters in the biology of malignant gliomas and highlight some of the unique aspects of these tumors that make them so devastating and difficult to treat.
Collapse
|
20
|
Rajkowska G, Stockmeier CA. Astrocyte pathology in major depressive disorder: insights from human postmortem brain tissue. Curr Drug Targets 2013; 14:1225-36. [PMID: 23469922 PMCID: PMC3799810 DOI: 10.2174/13894501113149990156] [Citation(s) in RCA: 436] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 02/22/2013] [Accepted: 02/26/2013] [Indexed: 02/07/2023]
Abstract
The present paper reviews astrocyte pathology in major depressive disorder (MDD) and proposes that reductions in astrocytes and related markers are key features in the pathology of MDD. Astrocytes are the most numerous and versatile of all types of glial cells. They are crucial to the neuronal microenvironment by regulating glucose metabolism, neurotransmitter uptake (particularly for glutamate), synaptic development and maturation and the blood brain barrier. Pathology of astrocytes has been consistently noted in MDD as well as in rodent models of depressive-like behavior. This review summarizes evidence from human postmortem tissue showing alterations in the expression of protein and mRNA for astrocyte markers such as glial fibrillary acidic protein (GFAP), gap junction proteins (connexin 40 and 43), the water channel aquaporin-4 (AQP4), a calcium-binding protein S100B and glutamatergic markers including the excitatory amino acid transporters 1 and 2 (EAAT1, EAAT2) and glutamine synthetase. Moreover, preclinical studies are presented that demonstrate the involvement of GFAP and astrocytes in animal models of stress and depressive-like behavior and the influence of different classes of antidepressant medications on astrocytes. In light of the various astrocyte deficits noted in MDD, astrocytes may be novel targets for the action of antidepressant medications. Possible functional consequences of altered expression of astrocytic markers in MDD are also discussed. Finally, the unique pattern of cell pathology in MDD, characterized by prominent reductions in the density of astrocytes and in the expression of their markers without obvious neuronal loss, is contrasted with that found in other neuropsychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Grazyna Rajkowska
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 N. State St., Box 127, Jackson, MS 39216-4505, USA.
| | | |
Collapse
|
21
|
Parpura V, Verkhratsky A. Astroglial amino acid-based transmitter receptors. Amino Acids 2013; 44:1151-8. [DOI: 10.1007/s00726-013-1458-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 01/02/2013] [Indexed: 10/27/2022]
|
22
|
Verkhratsky A, Reyes RC, Parpura V. TRP channels coordinate ion signalling in astroglia. Rev Physiol Biochem Pharmacol 2013; 166:1-22. [PMID: 23784619 DOI: 10.1007/112_2013_15] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Astroglial excitability is based on highly spatio-temporally coordinated fluctuations of intracellular ion concentrations, among which changes in Ca(2+) and Na(+) take the leading role. Intracellular signals mediated by Ca(2+) and Na(+) target numerous molecular cascades that control gene expression, energy production and numerous homeostatic functions of astrocytes. Initiation of Ca(2+) and Na(+) signals relies upon plasmalemmal and intracellular channels that allow fluxes of respective ions down their concentration gradients. Astrocytes express several types of TRP channels of which TRPA1 channels are linked to regulation of functional expression of GABA transporters, whereas TRPV4 channels are activated following osmotic challenges and are up-regulated in ischaemic conditions. Astrocytes also ubiquitously express several isoforms of TRPC channels of which heteromers assembled from TRPC1, 4 and/or 5 subunits that likely act as stretch-activated channels and are linked to store-operated Ca(2+) entry. The TRPC channels mediate large Na(+) fluxes that are associated with the endoplasmic reticulum Ca(2+) signalling machinery and hence coordinate Na(+) and Ca(2+) signalling in astroglia.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK,
| | | | | |
Collapse
|
23
|
Abstract
Astrocytes exhibit their excitability based on variations in cytosolic Ca(2+) levels, which leads to variety of signalling events. Only recently, however, intracellular fluctuations of more abundant cation Na(+) are brought in the limelight of glial signalling. Indeed, astrocytes possess several plasmalemmal molecular entities that allow rapid transport of Na(+) across the plasma membrane: (1) ionotropic receptors, (2) canonical transient receptor potential cation channels, (3) neurotransmitter transporters and (4) sodium-calcium exchanger. Concerted action of these molecules in controlling cytosolic Na(+) may complement Ca(2+) signalling to provide basis for complex bidirectional astrocyte-neurone communication at the tripartite synapse.
Collapse
|
24
|
Parpura V, Verkhratsky A. The astrocyte excitability brief: From receptors to gliotransmission. Neurochem Int 2012; 61:610-21. [DOI: 10.1016/j.neuint.2011.12.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2011] [Revised: 11/14/2011] [Accepted: 12/01/2011] [Indexed: 01/23/2023]
|
25
|
Abstract
Astroglial cells, due to their passive electrical properties, were long considered subservient to neurons and to merely provide the framework and metabolic support of the brain. Although astrocytes do play such structural and housekeeping roles in the brain, these glial cells also contribute to the brain's computational power and behavioural output. These more active functions are endowed by the Ca2+-based excitability displayed by astrocytes. An increase in cytosolic Ca2+ levels in astrocytes can lead to the release of signalling molecules, a process termed gliotransmission, via the process of regulated exocytosis. Dynamic components of astrocytic exocytosis include the vesicular-plasma membrane secretory machinery, as well as the vesicular traffic, which is governed not only by general cytoskeletal elements but also by astrocyte-specific IFs (intermediate filaments). Gliotransmitters released into the ECS (extracellular space) can exert their actions on neighbouring neurons, to modulate synaptic transmission and plasticity, and to affect behaviour by modulating the sleep homoeostat. Besides these novel physiological roles, astrocytic Ca2+ dynamics, Ca2+-dependent gliotransmission and astrocyte–neuron signalling have been also implicated in brain disorders, such as epilepsy. The aim of this review is to highlight the newer findings concerning Ca2+ signalling in astrocytes and exocytotic gliotransmission. For this we report on Ca2+ sources and sinks that are necessary and sufficient for regulating the exocytotic release of gliotransmitters and discuss secretory machinery, secretory vesicles and vesicle mobility regulation. Finally, we consider the exocytotic gliotransmission in the modulation of synaptic transmission and plasticity, as well as the astrocytic contribution to sleep behaviour and epilepsy.
Collapse
|
26
|
Chronic perinatal hypoxia reduces glutamate-aspartate transporter function in astrocytes through the Janus kinase/signal transducer and activator of transcription pathway. J Neurosci 2012; 31:17864-71. [PMID: 22159101 DOI: 10.1523/jneurosci.3179-11.2011] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The cellular and molecular mechanisms that govern the response of the perinatal brain to injury remain largely unexplored. We investigated the role of white matter astrocytes in a rodent model of diffuse white matter injury produced by exposing neonatal mice to chronic hypoxia-a paradigm that mimics brain injury in premature infants. We demonstrate the absence of reactive gliosis in the immature white matter following chronic hypoxia, as determined by astrocyte proliferation index and glial fibrillary acidic protein levels. Instead, Nestin expression in astrocytes is transiently increased, and the glial-specific glutamate transporters glutamate-aspartate transporter (GLAST) and glutamate transporter 1 (GLT-1) are reduced. Finally, we demonstrate that Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling-which is important in both astrocyte development and response to injury-is reduced in the white matter following hypoxia, as well as in primary astrocytes exposed to hypoxia in vitro. Hypoxia and JAK/STAT inhibition reduce glutamate transporter expression in astrocytes, but unlike hypoxia JAK/STAT inhibition downregulates GLAST expression without affecting GLT-1, as demonstrated in vitro by treatment with JAK inhibitor I and in vivo by treatment with the JAK/STAT inhibitor AG490 [(E)-2-cyano-3-(3,4-dihydrophenyl)-N-(phenylmethyl)-2-propenamide]. Our findings (1) demonstrate specific changes in astrocyte function after perinatal hypoxia, which might contribute to the particular pathogenesis of perinatal white matter injury, (2) provide evidence that at least part of these changes result from a disturbance of the JAK/STAT pathway by hypoxia, and (3) identify JAK/STAT signaling as a potential therapeutic target to restore normal GLAST expression and uptake of glutamate after perinatal brain injury.
Collapse
|
27
|
Illes P, Verkhratsky A, Burnstock G, Franke H. P2X receptors and their roles in astroglia in the central and peripheral nervous system. Neuroscientist 2011; 18:422-38. [PMID: 22013151 DOI: 10.1177/1073858411418524] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Astrocytes are a class of neural cells that control homeostasis at all levels of the central and peripheral nervous system. There is a bidirectional neuron-glia interaction via a number of extracellular signaling molecules, glutamate and ATP being the most widespread. ATP activates ionotropic P2X and metabotropic P2Y receptors, which operate in both neurons and astrocytes. Morphological, biochemical, and functional evidence indicates the expression of astroglial P2X(1/5) heteromeric and P2X(7) homomeric receptors, which mediate physiological and pathophysiological responses. Activation of P2X(1/5) receptors triggers rapid increase of intracellular Na(+) that initiates immediate cellular reactions, such as the depression of the glutamate transporter to keep high glutamate concentrations in the synaptic cleft, the activation of the local lactate shuttle to supply energy substrate to pre- and postsynaptic neuronal structures, and the reversal of the Na(+)/Ca(2+) exchange resulting in additional Ca(2+) entry. The consequences of P2X(7) receptor activation are mostly but not exclusively mediated by the entry of Ca(2+) and result in reorganization of the cytoskeleton, inflammation, apoptosis/necrosis, and proliferation, usually at a prolonged time scale. Thus, astroglia detect by P2X(1/5) and P2X(7) receptors both physiological concentrations of ATP secreted from presynaptic nerve terminals and also much higher concentrations of ATP attained under pathological conditions.
Collapse
Affiliation(s)
- Peter Illes
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, University of Leipzig, Germany.
| | | | | | | |
Collapse
|
28
|
Abstract
Demyelinating disorders of the central nervous system are among the most crippling neurological diseases affecting patients at various stages of life. In the most prominent demyelinating disease, multiple sclerosis, the regeneration of myelin sheaths often fails due to a default of the resident stem/precursor cells (oligodendrocyte precursor cells) to differentiate into mature myelin forming cells. Significant advances have been made in our understanding of the molecular and cellular processes involved in remyelination. Furthermore, important insight has been gained from studies investigating the interaction of stem/precursor cells with the distinct environment of demyelinating lesions. These suggest that successful regeneration depends on a signalling environment conducive to remyelination, which is provided in the context of acute inflammation. However, multiple sclerosis lesions also contain factors that inhibit the differentiation of oligodendrocyte precursor cells into myelinating oligodendrocytes. The pattern by which remyelination inducers and inhibitors are expressed in multiple sclerosis lesions may determine a window of opportunity during which oligodendrocyte precursor cells can successfully differentiate. As the first molecules aiming at promoting remyelination are about to enter clinical trials, this review critically evaluates recent advances in our understanding of the biology of oligodendrocyte precursor cells and of the stage-dependent molecular pathology of multiple sclerosis lesions relevant to the regeneration of myelin sheaths. We propose a model that may help to provide cues for how remyelination can be therapeutically enhanced in clinical disease.
Collapse
Affiliation(s)
- Mark R Kotter
- Department of Clinical Neurosciences, MRC Centre for Stem Cells and Regenerative Medicine, University of Cambridge, Addenbrooke's Hospital, Box 167, Hills Road, Cambridge CB22QQ, UK.
| | | | | |
Collapse
|
29
|
Vandenbroeck K, Comabella M. Single-nucleotide polymorphisms in response to interferon-beta therapy in multiple sclerosis. J Interferon Cytokine Res 2011; 30:727-32. [PMID: 20836710 DOI: 10.1089/jir.2010.0085] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Interferon-beta (IFN-β) is one of the main first-line disease-modifying drugs indicated for the treatment of multiple sclerosis (MS). The drug exhibits only limited effectiveness, and does not produce clinical benefits in around 20%-50% of patients. The availability of biomarkers would be beneficial for identification of patients at high risk of treatment failure, before initiation of therapy. Over the last 5 years, the search for such biomarkers has intensified and various promising candidates have been uncovered. Here, we review the main attempts undertaken to identify polymorphic variants associated with response to IFN-β therapy in MS by means of candidate gene approaches and whole-genome association scans. Despite substantial progress made in the field, there is still a long way to go before biomarker discoveries can be incorporated into clinical practice to predict IFN-β-responder status in MS patients.
Collapse
Affiliation(s)
- Koen Vandenbroeck
- Neurogenomiks Group, Universidad del País Vasco (UPV/EHU), Leioa, Spain.
| | | |
Collapse
|
30
|
Goubard V, Fino E, Venance L. Contribution of astrocytic glutamate and GABA uptake to corticostriatal information processing. J Physiol 2011; 589:2301-19. [PMID: 21486792 DOI: 10.1113/jphysiol.2010.203125] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The astrocytes, active elements of the tripartite synapse, remove most of the neurotransmitter that spills over the synaptic cleft. Neurotransmitter uptake operated by astrocytes contributes to the strength and timing of synaptic inputs. The striatum, the main input nucleus of basal ganglia, extracts pertinent cortical signals from the background noise and relays cortical information toward basal ganglia output structures. We investigated the role of striatal astrocytic uptake in the shaping of corticostriatal transmission.We performed dual patch-clamp recordings of striatal output neuron (the medium-sized spiny neurons, MSNs)–astrocyte pairs while stimulating the somatosensory cortex. Cortical activity evoked robust synaptically activated transporter-mediated currents (STCs) in 78% of the recorded astrocytes. STCs originated equally from the activities of glutamate transporters and GABA transporters (GATs). Astrocytic STCs reflected here a presynaptic release of neurotransmitters. STCs displayed a large magnitude associated with fast kinetics, denoting an efficient neurotransmitter clearance at the corticostriatal pathway. Inhibition of glutamate transporters type-1 (GLT-1) and GATs decreased the corticostriatal synaptic transmission, through, respectively, desensitization of AMPA receptors and activation of GABAA receptor. STCs displayed a bidirectional short-term plasticity (facilitation for paired-pulse intervals less than 100 ms and depression up to 1 s).We report a genuine facilitation of STCs for high-frequency cortical activity, which could strengthen the detection properties of cortical activity operated by MSNs. MSN EPSCs showed a triphasic short-term plasticity, which was modified by the blockade of GLT-1 or GATs. We show here that neurotransmitter uptake by astrocytes plays a key role in the corticostriatal information processing.
Collapse
Affiliation(s)
- Valérie Goubard
- Dynamic and Pathophysiology of Neuronal Networks, INSERM U667, College de France, 75005 Paris, France
| | | | | |
Collapse
|
31
|
Palygin O, Lalo U, Verkhratsky A, Pankratov Y. Ionotropic NMDA and P2X1/5 receptors mediate synaptically induced Ca2+ signalling in cortical astrocytes. Cell Calcium 2011; 48:225-31. [PMID: 20926134 DOI: 10.1016/j.ceca.2010.09.004] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 09/05/2010] [Accepted: 09/07/2010] [Indexed: 10/19/2022]
Abstract
Local, global and propagating calcium (Ca(2+)) signals provide the substrate for glial excitability. Here we analyse Ca(2+) permeability of NMDA and P2X(1/5) receptors expressed in cortical astrocytes and provide evidence that activation of these receptors trigger astroglial Ca(2+) signals when stimulated by either endogenous agonists or by synaptic release of neurotransmitters. The Ca(2+) permeability of the ionotropic receptors was determined by reversal potential shift analysis; the permeability ratio P(Ca)/P(K) was 3.1 for NMDA receptors and 2.2 for P2X(1/5) receptors. Selective stimulation of ionotropic receptors (with NMDA and α,β-methyleneATP) in freshly isolated cortical astrocytes induced ion currents associated with transient increases in cytosolic Ca(2+) concentration ([Ca(2+)](i)). Stimulation of neuronal afferents in cortical slices triggered glial synaptic currents and [Ca(2+)](i) responses, which were partially blocked by selective antagonists of NMDA (D-AP5 and UBP141) and P2X(1/5) (NF449) receptors. We conclude that ionotropic receptors contribute to astroglial Ca(2+) signalling and may provide a specific mechanism for fast neuronal-glial signalling at the synaptic level.
Collapse
Affiliation(s)
- Oleg Palygin
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| | | | | | | |
Collapse
|
32
|
Abstract
Astrocytes constitute a major group of glial cells which were long regarded as passive elements, fulfilling nutritive and structural functions for neurons. Calcium rise in astrocytes propagating to neurons was the first demonstration of direct interaction between the two cell types. Since then, calcium has been widely used, not only as an indicator of astrocytic activity but also as a stimulator switch to control astrocyte physiology. As a result, astrocytes have been elevated from auxiliaries to neurons, to cells involved in processing synaptic information. Curiously, while there is evidence that astrocytes play an important role in synaptic plasticity, the data relating to calcium's pivotal role are inconsistent. In this review, we will detail the various mechanisms of calcium flux in astrocytes, then briefly present the calcium-dependent mechanisms of gliotransmitter release. Finally, we will discuss the role of calcium in plasticity and present alternative explanations that could reconcile the conflicting results published recently.
Collapse
|
33
|
Parpura V, Grubišić V, Verkhratsky A. Ca(2+) sources for the exocytotic release of glutamate from astrocytes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1813:984-91. [PMID: 21118669 DOI: 10.1016/j.bbamcr.2010.11.006] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Revised: 11/07/2010] [Accepted: 11/10/2010] [Indexed: 01/26/2023]
Abstract
Astrocytes can exocytotically release the gliotransmitter glutamate from vesicular compartments. Increased cytosolic Ca(2+) concentration is necessary and sufficient for this process. The predominant source of Ca(2+) for exocytosis in astrocytes resides within the endoplasmic reticulum (ER). Inositol 1,4,5-trisphosphate and ryanodine receptors of the ER provide a conduit for the release of Ca(2+) to the cytosol. The ER store is (re)filled by the store-specific Ca(2+)-ATPase. Ultimately, the depleted ER is replenished by Ca(2+) which enters from the extracellular space to the cytosol via store-operated Ca(2+) entry; the TRPC1 protein has been implicated in this part of the astrocytic exocytotic process. Voltage-gated Ca(2+) channels and plasma membrane Na(+)/Ca(2+) exchangers are additional means for cytosolic Ca(2+) entry. Cytosolic Ca(2+) levels can be modulated by mitochondria, which can take up cytosolic Ca(2+) via the Ca(2+) uniporter and release Ca(2+) into cytosol via the mitochondrial Na(+)/Ca(2+) exchanger, as well as by the formation of the mitochondrial permeability transition pore. The interplay between various Ca(2+) sources generates cytosolic Ca(2+) dynamics that can drive Ca(2+)-dependent exocytotic release of glutamate from astrocytes. An understanding of this process in vivo will reveal some of the astrocytic functions in health and disease of the brain. This article is part of a Special Issue entitled: 11th European Symposium on Calcium.
Collapse
Affiliation(s)
- Vladimir Parpura
- Department of Neurobiology, Center for Glial Biology in Medicine, Civitan International Research Center, Atomic Force Microscopy and Nanotechnology Laboratories, and Evelyn F. McKnight Brain Institute, University of Alabama, Birmingham 35294-0021, USA.
| | | | | |
Collapse
|
34
|
Verkhratsky A. Physiology of neuronal–glial networking. Neurochem Int 2010; 57:332-43. [DOI: 10.1016/j.neuint.2010.02.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Revised: 01/05/2010] [Accepted: 02/01/2010] [Indexed: 10/19/2022]
|
35
|
Ionotropic receptors in neuronal-astroglial signalling: what is the role of "excitable" molecules in non-excitable cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1813:992-1002. [PMID: 20869992 DOI: 10.1016/j.bbamcr.2010.09.007] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2010] [Revised: 09/13/2010] [Accepted: 09/16/2010] [Indexed: 01/02/2023]
Abstract
Astroglial cells were long considered to serve merely as the structural and metabolic supporting cast and scenery against which the shining neurones perform their illustrious duties. Relatively recent evidence, however, indicates that astrocytes are intimately involved in many of the brain's functions. Astrocytes possess a diverse assortment of ionotropic transmitter receptors, which enable these glial cells to respond to many of the same signals that act on neurones. Ionotropic receptors mediate neurone-driven signals to astroglial cells in various brain areas including neocortex, hippocampus and cerebellum. Activation of ionotropic receptors trigger rapid signalling events in astroglia; these events, represented by local Ca(2+) or Na(+) signals provide the mechanism for fast neuronal-glial signalling at the synaptic level. Since astrocytes can detect chemical transmitters that are released from neurones and can release their own extracellular signals, gliotransmitters, they are intricately involved in homocellular and heterocellular signalling mechanisms in the nervous system. This article is part of a Special Issue entitled: 11th European Symposium on Calcium.
Collapse
|
36
|
Abstract
Our view of astrocytes in the operation of the brain is changing dramatically over the last 3 decades. Astroglial calcium excitability controls the release of gliotransmitters, which can occur at the tripartite synapse. Astrocytes not only modulate synaptic transmission by releasing and taking up transmitters, but also receiving neuronal signals that act upon astrocytic plasma membrane receptors. This process represents the bidirectional neurone-glia communication. Additionally, astrocytes play role in the regulation of blood flow as well as ion and water homeostasis. Many of the brain dysfunctions are primary astropathies, including hepatic encephalopathy and Alexander disease, while other brain malfunctions, such as epilepsy and Alzheimer disease, may have substantial astrocytic contribution. Thus, these star-shaped cells by their roles in (patho)physiology of the brain seem to live up to the expectation one can have from their given name - astrocyte.
Collapse
|
37
|
Lim CK, Brew BJ, Sundaram G, Guillemin GJ. Understanding the roles of the kynurenine pathway in multiple sclerosis progression. Int J Tryptophan Res 2010; 3:157-67. [PMID: 22084596 PMCID: PMC3195238 DOI: 10.4137/ijtr.s4294] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The kynurenine pathway (KP) is a major degradative pathway of tryptophan ultimately leading to the production of nicotinamide adenine dinucleotide (NAD+) and is also one of the major regulatory mechanisms of the immune response. The KP is known to be involved in several neuroinflammatory disorders including Alzheimer’s disease, amyotrophic lateral sclerosis, AIDS dementia complex, Parkinson’s disease, schizophrenia, Huntington’s disease and brain tumours. However, the KP remains a relatively new topic for the field of multiple sclerosis (MS). Over the last 2–3 years, some evidence has progressively emerged suggesting that the KP is likely to be involved in the pathogenesis of autoimmune diseases especially MS. Some KP modulators are already in clinical trials for other inflammatory diseases and would potentially provide a new and important therapeutic strategy for MS patients. This review summarizes the known relationships between the KP and MS.
Collapse
Affiliation(s)
- Chai K Lim
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney 2052, Australia
| | | | | | | |
Collapse
|
38
|
Verkhratsky A, Parpura V, Rodríguez JJ. Where the thoughts dwell: the physiology of neuronal-glial "diffuse neural net". ACTA ACUST UNITED AC 2010; 66:133-51. [PMID: 20546785 DOI: 10.1016/j.brainresrev.2010.05.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Revised: 05/11/2010] [Accepted: 05/17/2010] [Indexed: 02/07/2023]
Abstract
The mechanisms underlying the production of thoughts by exceedingly complex cellular networks that construct the human brain constitute the most challenging problem of natural sciences. Our understanding of the brain function is very much shaped by the neuronal doctrine that assumes that neuronal networks represent the only substrate for cognition. These neuronal networks however are embedded into much larger and probably more complex network formed by neuroglia. The latter, although being electrically silent, employ many different mechanisms for intercellular signalling. It appears that astrocytes can control synaptic networks and in such a capacity they may represent an integral component of the computational power of the brain rather than being just brain "connective tissue". The fundamental question of whether neuroglia is involved in cognition and information processing remains, however, open. Indeed, a remarkable increase in the number of glial cells that distinguishes the human brain can be simply a result of exceedingly high specialisation of the neuronal networks, which delegated all matters of survival and maintenance to the neuroglia. At the same time potential power of analogue processing offered by internally connected glial networks may represent the alternative mechanism involved in cognition.
Collapse
|
39
|
Cruz-Solis I, Zepeda RC, Ortiz S, Aguilera J, López-Bayghen E, Ortega A. Glutamate-dependent transcriptional control in Bergmann glia: Sox10 as a repressor. J Neurochem 2009; 109:899-910. [PMID: 19425178 DOI: 10.1111/j.1471-4159.2009.06017.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
UNLABELLED Glutamate (Glu) is the major excitatory transmitter in the vertebrate brain. Ligand-gated and G protein-coupled Glu receptors present in glial cells are presumably involved in neuronal function. Activation of Bergmann glial Glu receptors triggers a membrane to nuclei signaling cascade that regulates gene expression at the transcriptional and translational levels. Sry-related high-mobility group box (Sox10), a member of the conserved high-mobility group box transcription factor family is expressed in neural crest stem cells and in a subset of neural crest-derived lineages that include glial, but not neuronal cells. To gain insight into the role of Sox10 in Bergmann glial cells, we explored its expression and regulation. We demonstrate herein that Sox10 is expressed in Bergmann glial cells and that its DNA binding activity, mRNA, and protein levels as well as its transcriptional behavior augments upon the activation of metabotropic Glu receptors. Increase in Sox10-DNA complexes and Sox10 mRNA and protein levels were found upon exposure to Glu. Over-expression of Sox10 leads to transcriptional repression in reporter gene assays and in one of its target genes: the chick kainate binding protein gene. These findings add a new perspective into glial glutamatergic signaling and suggest the participation of Sox10 in cerebellar glutamatergic transactions. KEYWORDS Bergmann glial cells, glutamate, metabotropic glutamate receptors, signaling, Sox10, transcriptional control.
Collapse
Affiliation(s)
- Irma Cruz-Solis
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Apartado, México
| | | | | | | | | | | |
Collapse
|
40
|
Ouardouz M, Coderre E, Zamponi GW, Hameed S, Yin X, Trapp BD, Stys PK. Glutamate receptors on myelinated spinal cord axons: II. AMPA and GluR5 receptors. Ann Neurol 2009; 65:160-6. [PMID: 19224531 DOI: 10.1002/ana.21539] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Glutamate receptors, which play a major role in the physiology and pathology of central nervous system gray matter, are also involved in the pathophysiology of white matter. However, the cellular and molecular mechanisms responsible for excitotoxic damage to white matter elements are not fully understood. We explored the roles of AMPA and GluR5 kainate receptors in axonal Ca(2+) deregulation. METHODS Dorsal column axons were loaded with a Ca(2+) indicator and imaged in vitro using confocal microscopy. RESULTS Both AMPA and a GluR5 kainate receptor agonist increased intraaxonal Ca(2+) in myelinated rat dorsal column fibers. These responses were inhibited by selective antagonists of these receptors. The GluR5-mediated Ca(2+) increase was mediated by both canonical (ie, ionotropic) and noncanonical (metabotropic) signaling, dependent on a pertussis toxin-sensitive G protein/phospholipase C-dependent pathway, promoting Ca(2+) release from inositol triphosphate-dependent stores. In addition, the GluR5 response was reduced by intraaxonal NO scavengers. In contrast, GluR4 AMPA receptors operated via Ca(2+)-induced Ca(2+) release, dependent on ryanodine receptors, and unaffected by NO scavengers. Neither pathway depended on L-type Ca(2+) channels, in contrast with GluR6 kainate receptor action.1 Immunohistochemistry confirmed the presence of GluR4 and GluR5 clustered at the surface of myelinated axons; GluR5 coimmunoprecipitated with nNOS and often colocalized with neuronal nitric oxide synthase clusters on the internodal axon. INTERPRETATION Central myelinated axons express functional AMPA and GluR5 kainate receptors, and can directly respond to glutamate receptor agonists. These glutamate receptor-dependent signaling pathways promote an increase in intraaxonal Ca(2+) levels potentially contributing to axonal degeneration.
Collapse
Affiliation(s)
- Mohamed Ouardouz
- Ottawa Health Research Institute, University of Ottawa, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
41
|
Casley CS, Lakics V, Lee HG, Broad LM, Day TA, Cluett T, Smith MA, O'Neill MJ, Kingston AE. Up-regulation of astrocyte metabotropic glutamate receptor 5 by amyloid-β peptide. Brain Res 2009; 1260:65-75. [PMID: 19401173 DOI: 10.1016/j.brainres.2008.12.082] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2008] [Revised: 12/19/2008] [Accepted: 12/22/2008] [Indexed: 01/20/2023]
Abstract
The effects of amyloid-beta peptide (Aβ) on astrocyte responses to activation of mGlu5 receptors have been investigated using calcium imaging. Pre-incubation with Aβ1-40 peptide for up to 72 h produced a time- and concentration-dependent 2-4 fold enhancement in the magnitude of the intracellular calcium mobilization response to the group I metabotropic glutamate receptor agonist (S)-3,5-dihydroxyphenylglycine (DHPG). In contrast, pre-treatment with Aβ1-40 did not alter the calcium responses induced by other G protein coupled- or ion channel-receptors. Aβ 1-40-enhanced DHPG responses were blocked by the mGlu5 antagonist MPEP but not by inhibitors of voltage dependent calcium channels or by the AMPA/KA receptor antagonist CNQX. Up-regulation of mGlu5 coupled responses was associated with significant increases in astrocyte mGlu5 receptor-mRNA and-protein expression after preincubation with Aβ . The changes observed in vitro were consistent with results obtained from human Alzheimer's disease (AD) patients.Immunostaining for mGlu5 receptors was increased on astrocytes which were colocalized with Aβ plaques in hippocampal tissue from AD patients compared to age-matched controls. These results suggest that modulation of mGlu5 receptors in astrocytes could be an important mechanism in determining the progression of pathology in AD.
Collapse
Affiliation(s)
- Christopher S Casley
- Eli Lilly & Company Limited, Lilly Research Centre, Erl Wood Manor, Windlesham, Surrey, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Karram K, Goebbels S, Schwab M, Jennissen K, Seifert G, Steinhäuser C, Nave KA, Trotter J. NG2-expressing cells in the nervous system revealed by the NG2-EYFP-knockin mouse. Genesis 2008; 46:743-57. [DOI: 10.1002/dvg.20440] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
43
|
Durand D, Pampillo M, Caruso C, Lasaga M. Role of metabotropic glutamate receptors in the control of neuroendocrine function. Neuropharmacology 2008; 55:577-83. [PMID: 18616955 DOI: 10.1016/j.neuropharm.2008.06.022] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 05/23/2008] [Accepted: 06/12/2008] [Indexed: 11/25/2022]
Abstract
Glutamate exerts its effects through binding and activation of two classes of specific receptors: ionotropic (iGluRs) and metabotropic (mGluRs). Group I mGluR includes mGluR1 and mGluR5 subtypes, group II includes mGluR2 and mGluR3 subtypes and group III includes the subtypes mGluR 4, 6, 7 and 8. Glutamate and its receptors are found in all key hypothalamic areas critically involved in reproduction and neuroendocrine function. To date, considerable data support an important role for iGluRs in the control of neuroendocrine function; however, the role of mGluRs as regulators of hypothalamic-pituitary function has not been clearly elucidated. mGluRs could be exerting a fine tune on the release of hypothalamic factors that regulate hormone release such as Substance P, GABA, alpha-MSH and CRH. Group II mGluR exert a direct inhibitory effect on anterior pituitary prolactin and GH secretion. Moreover, some group II mGluR agonists, like LY 354,740 and LY 379,268, can modulate PRL secretion from the anterior pituitary through their actions as dopamine receptor agonists. Evidence suggests a role for group III mGluR subtypes in stress-related behavioral disorders. Several reports indicate that selective ligands for mGluR subtypes have potential for the treatment of a wide variety of neurological and psychiatric disorders, including depression, anxiety disorders, schizophrenia, epilepsy and Alzheimer's disease among others. Since converging lines of evidence suggest a role for mGluRs subtypes in neuroendocrine regulation of hormone secretion, mGluRs neuroendocrine actions must be taken in consideration to insure proper treatment of these diseases. Moreover, discovery of selective agonists provides an opportunity to investigate the physiological role of mGluR subtypes and to directly test the neuroendocrine actions of mGluRs. Finally, mGluRs selective agonists may have an impact in the treatment of conditions involving chronic stress, such as depression and anxiety disorders, since they regulate neuroendocrine stress circuits involving the HPA axis and stress-sensitive hormones such as oxytocin and prolactin. This review aims to provide a survey of our current understanding of the effects of mGluR activation on neuroendocrine function.
Collapse
Affiliation(s)
- Daniela Durand
- Research Center in Reproduction, School of Medicine, University of Buenos Aires, Paraguay 2155, Piso 10, 1121 Buenos Aires, Argentina
| | | | | | | |
Collapse
|
44
|
Cenci C, Barzotti R, Galeano F, Corbelli S, Rota R, Massimi L, Di Rocco C, O'Connell MA, Gallo A. Down-regulation of RNA editing in pediatric astrocytomas: ADAR2 editing activity inhibits cell migration and proliferation. J Biol Chem 2008; 283:7251-60. [PMID: 18178553 DOI: 10.1074/jbc.m708316200] [Citation(s) in RCA: 151] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Since alterations in post-transcriptional events can contribute to the appearance and/or progression of cancer, we investigated whether RNA editing, catalyzed by the ADAR (adenosine deaminases that act on RNA) enzymes, is altered in pediatric astrocytomas. We find a decrease in ADAR2 editing activity that seems to correlate with the grade of malignancy in children. Despite the loss of ADAR2 editing activity in tumor tissues, the high grade astrocytomas do not exhibit alterations in ADAR2 expression when compared with their specific control tissues. However, high expression levels of ADAR1 and ADAR3 were found in tumors when compared with normal tissues dissected in the same area of the brain. We reintroduced either ADAR2 or the inactive version of ADAR2 in three astrocytoma cell lines (U118, A172, U87). The "reverted" editing status is necessary and sufficient for a significant decrease in cell malignant behavior as measured by proliferation, cell cycle, and migration assays. We show that elevated levels of ADAR1, as found in astrocytomas, do indeed interfere with ADAR2 specific editing activity. Furthermore, we show that the endogenous ADAR1 can form heterodimers with ADAR2 in astrocytes.
Collapse
Affiliation(s)
- Caterina Cenci
- RNA editing Laboratory, Ospedale Pediatrico Bambino Gesù Research Institute, Piazza S. Onofrio 4, 00165 Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Janáky R, Shaw CA, Oja SS, Saransaari P. Taurine release in developing mouse hippocampus is modulated by glutathione and glutathione derivatives. Amino Acids 2008; 34:75-80. [PMID: 17701096 PMCID: PMC2814815 DOI: 10.1007/s00726-007-0587-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2007] [Accepted: 07/08/2007] [Indexed: 11/24/2022]
Abstract
Glutathione (reduced form GSH and oxidized form GSSG) constitutes an important defense against oxidative stress in the brain, and taurine is an inhibitory neuromodulator particularly in the developing brain. The effects of GSH and GSSG and glycylglycine, gamma-glutamylcysteine, cysteinylglycine, glycine and cysteine on the release of [(3)H]taurine evoked by K+-depolarization or the ionotropic glutamate receptor agonists glutamate, kainate, 2-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) and N-methyl-D-aspartate (NMDA) were now studied in slices from the hippocampi from 7-day-old mouse pups in a perfusion system. All stimulatory agents (50 mM K(+), 1 mM glutamate, 0.1 mM kainate, 0.1 mM AMPA and 0.1 mM NMDA) evoked taurine release in a receptor-mediated manner. Both GSH and GSSG significantly inhibited the release evoked by 50 mM K+. The release induced by AMPA and glutamate was also inhibited, while the kainate-evoked release was significantly activated by both GSH and GSSG. The NMDA-evoked release proved the most sensitive to modulation: L-Cysteine and glycine enhanced the release in a concentration-dependent manner, whereas GSH and GSSG were inhibitory at low (0.1 mM) but not at higher (1 or 10 mM) concentrations. The release evoked by 0.1 mM AMPA was inhibited by gamma-glutamylcysteine and cysteinylglycine, whereas glycylglycine had no effect. The 0.1 mM NMDA-evoked release was inhibited by glycylglycine and gamma-glutamylcysteine. In turn, cysteinylglycine inhibited the NMDA-evoked release at 0.1 mM, but was inactive at 1 mM. Glutathione exhibited both enhancing and attenuating effects on taurine release, depending on the glutathione concentration and on the agonist used. Both glutathione and taurine act as endogenous neuroprotective effectors during early postnatal life.
Collapse
Affiliation(s)
- R Janáky
- Brain Research Center, University of Tampere Medical School, Tampere, Finland
| | | | | | | |
Collapse
|
46
|
A modelling approach to explore some hypotheses of the failure of neuroprotective trials in ischemic stroke patients. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2007; 97:60-78. [PMID: 18076975 DOI: 10.1016/j.pbiomolbio.2007.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Ischemic stroke is the third cause of death in industrialised countries, but no satisfactory treatment is currently available. The hundreds of neuroprotective drugs developed to block the ischemic cascade gave very promising results in animal models but the clinical trials performed with these drugs showed no beneficial effects in stroke patients. Many hypotheses were advanced to explain this discrepancy, among which the morphological and functional differences between human and rodent brains. This discrepancy could be partly due to the differences in white matter and glial cell proportions between human and rodent brains. In order to test this hypothesis, we built a mathematical model of the main early pathophysiological mechanisms of stroke in rodent and in human brains. This model is a two-scale model and relies on a set of ordinary differential equations. We built two versions of this model (for human and rodent brains) differing in their white matter and glial cell proportions. Then, we carried out in silico experiments with various neuroprotective drugs. The simulation results obtained with a sodium channel blocker show that the proportion of penumbra recovery is much higher in rodent than in human brain and the results are similar with some other neuroprotective drugs tested during phase III trials. This in silico investigation suggests that the proportions of glial cells and white matter have an influence on neuroprotective drug efficacy. It reinforces the hypothesis that histological and morphological differences between rodent and human brains can partly explain the failure of these agents in clinical trials.
Collapse
|
47
|
Girard N, Confort-Gouny S, Schneider J, Chapon F, Viola A, Pineau S, Combaz X, Cozzone P. Neuroimaging of neonatal encephalopathies. J Neuroradiol 2007; 34:167-82. [PMID: 17590440 DOI: 10.1016/j.neurad.2007.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Neonatal brain disorders consist of a wide chapter including brain malformations, hypoxic-ischemic encephalopathy (HIE), intracranial infections, perinatal trauma and metabolic encephalopathy. We will focus here on HIE, intracranial infections (especially materno-fetal infection with or without prolonged and/or premature rupture of membranes) and metabolic encephalopathy, those three conditions being the most frequent so far in our experience. Neonatal stroke is also analyzed. Moreover minor perinatal events might be superimposed on an already damaged (infective, edematous, metabolically abnormal or maldeveloped) brain, highlighting the main role and potential benefits of neuroimaging during the neonatal period. The different methods of brain imaging are thus reported with their advantages and disadvantages.
Collapse
Affiliation(s)
- N Girard
- Department of Neuroradiology Diagnostique and Interventionnelle, hôpital Timone, université de la Méditerranée, 264 rue Saint-Pierre, 13005 Marseille, France.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Rajkowska G, Miguel-Hidalgo JJ. Gliogenesis and glial pathology in depression. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2007; 6:219-33. [PMID: 17511618 PMCID: PMC2918806 DOI: 10.2174/187152707780619326] [Citation(s) in RCA: 447] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recent research has changed the perception of glia from being no more than silent supportive cells of neurons to being dynamic partners participating in brain metabolism and communication between neurons. This discovery of new glial functions coincides with growing evidence of the involvement of glia in the neuropathology of mood disorders. Unanticipated reductions in the density and number of glial cells are reported in fronto-limbic brain regions in major depression and bipolar illness. Moreover, age-dependent decreases in the density of glial fibrillary acidic protein (GFAP) - immunoreactive astrocytes and levels of GFAP protein are observed in the prefrontal cortex of younger depressed subjects. Since astrocytes participate in the uptake, metabolism and recycling of glutamate, we hypothesize that an astrocytic deficit may account for the alterations in glutamate/GABA neurotransmission in depression. Reductions in the density and ultrastructure of oligodendrocytes are also detected in the prefrontal cortex and amygdala in depression. Pathological changes in oligodendrocytes may be relevant to the disruption of white matter tracts in mood disorders reported by diffusion tensor imaging. Factors such as stress, excess of glucocorticoids, altered gene expression of neurotrophic factors and glial transporters, and changes in extracellular levels of neurotransmitters released by neurons may modify glial cell number and affect the neurophysiology of depression. Therefore, we will explore the role of these events in the possible alteration of glial number and activity, and the capacity of glia as a promising new target for therapeutic medications. Finally, we will consider the temporal relationship between glial and neuronal cell pathology in depression.
Collapse
Affiliation(s)
- G Rajkowska
- Department of Psychiatry, University of Mississippi Medical Center, Jackson, MS 39216-4505, USA.
| | | |
Collapse
|
49
|
Kukley M, Capetillo-Zarate E, Dietrich D. Vesicular glutamate release from axons in white matter. Nat Neurosci 2007; 10:311-20. [PMID: 17293860 DOI: 10.1038/nn1850] [Citation(s) in RCA: 367] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2006] [Accepted: 01/16/2007] [Indexed: 12/11/2022]
Abstract
Vesicular release of neurotransmitter is the universal output signal of neurons in the brain. It is generally believed that fast transmitter release is restricted to nerve terminals that contact postsynaptic cells in the gray matter. Here we show in the rat brain that the neurotransmitter glutamate is also released at discrete sites along axons in white matter in the absence of neurons and nerve terminals. The propagation of single action potentials along axons leads to rapid vesicular release of glutamate, which is detected by ionotropic glutamate receptors on local oligodendrocyte precursor cells. Axonal release of glutamate is reliable, involves highly localized calcium microdomain signaling and is strongly calcium cooperative, similar to vesicle fusion at synapses. This axonal transmitter release represents a widespread mechanism for high-fidelity, activity-dependent signaling at the axon-glia interface in white matter.
Collapse
Affiliation(s)
- Maria Kukley
- Experimental Neurophysiology, Department of Neurosurgery, University Clinic Bonn, Germany.
| | | | | |
Collapse
|
50
|
Karavanova I, Vasudevan K, Cheng J, Buonanno A. Novel regional and developmental NMDA receptor expression patterns uncovered in NR2C subunit-beta-galactosidase knock-in mice. Mol Cell Neurosci 2007; 34:468-80. [PMID: 17276696 PMCID: PMC1855159 DOI: 10.1016/j.mcn.2006.12.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2006] [Accepted: 12/05/2006] [Indexed: 11/25/2022] Open
Abstract
NMDA receptor "knock-in" mice were generated by inserting the nuclear beta-galactosidase reporter at the NR2C subunit translation initiation site. Novel cell types and dynamic patterns of NR2C expression were identified using these mice, which were unnoticed before because reagents that specifically recognize NR2C-containing receptors are non-existent. We identified a transition zone from NR2C-expressing neurons to astrocytes in an area connecting the retrosplenial cortex and hippocampus. We demonstrate that NR2C is expressed in a subset of S100beta-positive/GFAP-negative glial cells in the striatum, olfactory bulb and cerebral cortex. We also demonstrate novel areas of neuronal expression such as retrosplenial cortex, thalamus, pontine and vestibular nuclei. In addition, we show that during cerebellar development NR2C is expressed in transient caudal-rostral gradients and parasagittal bands in subsets of granule cells residing in the internal granular layer, further demonstrating heterogeneity of granule neurons. These results point to novel functions of NR2C-containing NMDA receptors.
Collapse
Affiliation(s)
| | | | | | - Andres Buonanno
- Correspondence addressed to: Andres Buonanno, Ph.D. Section on Molecular Neurobiology, Chief, Bldg 35, Room 2C-1000, 35 Lincoln Drive, National Institutes of Health, Bethesda, Maryland 20892, USA, e-mail:
| |
Collapse
|