1
|
Millet LJ, Jain A, Gillette MU. Less Is More: Oligomer Extraction and Hydrothermal Annealing Increase PDMS Adhesion Forces for Materials Studies and for Biology-Focused Microfluidic Applications. MICROMACHINES 2023; 14:214. [PMID: 36677275 PMCID: PMC9866318 DOI: 10.3390/mi14010214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/30/2022] [Accepted: 01/07/2023] [Indexed: 06/17/2023]
Abstract
Cues in the micro-environment are key determinants in the emergence of complex cellular morphologies and functions. Primary among these is the presence of neighboring cells that form networks. For high-resolution analysis, it is crucial to develop micro-environments that permit exquisite control of network formation. This is especially true in cell science, tissue engineering, and clinical biology. We introduce a new approach for assembling polydimethylsiloxane (PDMS)-based microfluidic environments that enhances cell network formation and analyses. We report that the combined processes of PDMS solvent-extraction and hydrothermal annealing create unique conditions that produce high-strength bonds between solvent-extracted PDMS (E-PDMS) and glass-properties not associated with conventional PDMS. Extraction followed by hydrothermal annealing removes unbound oligomers, promotes polymer cross-linking, facilitates covalent bond formation with glass, and retains the highest biocompatibility. Herein, our extraction protocol accelerates oligomer removal from 5 to 2 days. Resulting microfluidic platforms are uniquely suited for cell-network studies owing to high adhesion forces, effectively corralling cellular extensions and eliminating harmful oligomers. We demonstrate the simple, simultaneous actuation of multiple microfluidic domains for invoking ATP- and glutamate-induced Ca2+ signaling in glial-cell networks. These E-PDMS modifications and flow manipulations further enable microfluidic technologies for cell-signaling and network studies as well as novel applications.
Collapse
Affiliation(s)
- Larry J. Millet
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- The Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Biosciences Division, Oak Ridge National Laboratory, One Bethel Valley Road, Oak Ridge, TN 37831, USA
- The Center for Environmental Biotechnology, University of Tennessee Knoxville, Knoxville, TN 37996, USA
| | - Anika Jain
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- The Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Martha U. Gillette
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- The Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Neuroscience Program, Beckman Institute for Advanced Science & Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
2
|
Role of Microglia and Astrocytes in Alzheimer’s Disease: From Neuroinflammation to Ca2+ Homeostasis Dysregulation. Cells 2022; 11:cells11172728. [PMID: 36078138 PMCID: PMC9454513 DOI: 10.3390/cells11172728] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia worldwide, with a complex, poorly understood pathogenesis. Cerebral atrophy, amyloid-β (Aβ) plaques, and neurofibrillary tangles represent the main pathological hallmarks of the AD brain. Recently, neuroinflammation has been recognized as a prominent feature of the AD brain and substantial evidence suggests that the inflammatory response modulates disease progression. Additionally, dysregulation of calcium (Ca2+) homeostasis represents another early factor involved in the AD pathogenesis, as intracellular Ca2+ concentration is essential to ensure proper cellular and neuronal functions. Although growing evidence supports the involvement of Ca2+ in the mechanisms of neurodegeneration-related inflammatory processes, scant data are available on its contribution in microglia and astrocytes functioning, both in health and throughout the AD continuum. Nevertheless, AD-related aberrant Ca2+ signalling in astrocytes and microglia is crucially involved in the mechanisms underpinning neuroinflammatory processes that, in turn, impact neuronal Ca2+ homeostasis and brain function. In this light, we attempted to provide an overview of the current understanding of the interactions between the glia cells-mediated inflammatory responses and the molecular mechanisms involved in Ca2+ homeostasis dysregulation in AD.
Collapse
|
3
|
Astrocyte networks modulate respiration – sniffing glue. Respir Physiol Neurobiol 2019; 265:3-8. [DOI: 10.1016/j.resp.2018.06.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/17/2018] [Accepted: 06/29/2018] [Indexed: 12/11/2022]
|
4
|
Verkhratsky A, Chvátal A. NMDA Receptors in Astrocytes. Neurochem Res 2019; 45:122-133. [DOI: 10.1007/s11064-019-02750-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 01/22/2019] [Accepted: 02/05/2019] [Indexed: 12/12/2022]
|
5
|
McClain JL, Gulbransen BD. The acute inhibition of enteric glial metabolism with fluoroacetate alters calcium signaling, hemichannel function, and the expression of key proteins. J Neurophysiol 2016; 117:365-375. [PMID: 27784805 DOI: 10.1152/jn.00507.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 10/25/2016] [Indexed: 11/22/2022] Open
Abstract
Glia play key roles in the regulation of neurotransmission in the nervous system. Fluoroacetate (FA) is a metabolic poison widely used to study glial functions by disrupting the tricarboxylic acid cycle enzyme aconitase. Despite the widespread use of FA, the effects of FA on essential glial functions such as calcium (Ca2+) signaling and hemichannel function remain unknown. Therefore, our goal was to assess specifically the impact of FA on essential glial cell functions that are involved with neurotransmission in the enteric nervous system. To this end, we generated a new optogenetic mouse model to study specifically the effects of FA on enteric glial Ca2+ signaling by crossing PC::G5-tdTomato mice with Sox10::creERT2 mice. FA did not change the peak glial Ca2+ response when averaged across all glia within a ganglion. However, FA decreased the percent of responding glia by 30% (P < 0.05) and increased the peak Ca2+ response of the glial cells that still exhibited a response by 26% (P < 0.01). Disruption of Ca2+ signaling with FA impaired the activity-dependent uptake of ethidium bromide through connexin-43 (Cx43) hemichannels (P < 0.05) but did not affect baseline Cx43-dependent dye uptake. FA did not cause overt glial or neurodegeneration, but glial cells significantly increased glial fibrillary acid protein by 56% (P < 0.05) following treatment with FA. Together, these data show that the acute impairment of glial metabolism with FA causes key changes in glial functions associated with their roles in neurotransmission and phenotypic changes indicative of reactive gliosis. NEW & NOTEWORTHY Our study shows that the acute impairment of enteric glial metabolism with fluoroacetate (FA) alters specific glial functions that are associated with the modification of neurotransmission in the gut. These include subtle changes to glial agonist-evoked calcium signaling, the subsequent disruption of connexin-43 hemichannels, and changes in protein expression that are consistent with a transition to reactive glia. These changes in glial function offer a mechanistic explanation for the effects of FA on peripheral neuronal networks.
Collapse
Affiliation(s)
- Jonathon L McClain
- Department of Physiology, Michigan State University, East Lansing, Michigan; and
| | - Brian D Gulbransen
- Department of Physiology, Michigan State University, East Lansing, Michigan; and .,Neuroscience Program, Michigan State University, East Lansing, Michigan
| |
Collapse
|
6
|
Astroglial calcium signalling in Alzheimer's disease. Biochem Biophys Res Commun 2016; 483:1005-1012. [PMID: 27545605 DOI: 10.1016/j.bbrc.2016.08.088] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 08/15/2016] [Indexed: 12/14/2022]
Abstract
Neuroglial contribution to Alzheimer's disease (AD) is pathologically relevant and highly heterogeneous. Reactive astrogliosis and activation of microglia contribute to neuroinflammation, whereas astroglial and oligodendroglial atrophy affect synaptic transmission and underlie the overall disruption of the central nervous system (CNS) connectome. Astroglial function is tightly integrated with the intracellular ionic signalling mediated by complex dynamics of cytosolic concentrations of free Ca2+ and Na+. Astroglial ionic signalling is mediated by plasmalemmal ion channels, mainly associated with ionotropic receptors, pumps and solute carrier transporters, and by intracellular organelles comprised of the endoplasmic reticulum and mitochondria. The relative contribution of these molecular cascades/organelles can be plastically remodelled in development and under environmental stress. In AD astroglial Ca2+ signalling undergoes substantial reorganisation due to an abnormal regulation of expression of Ca2+ handling molecular cascades.
Collapse
|
7
|
Rapôso C, Björklund U, Kalapothakis E, Biber B, Alice da Cruz-Höfling M, Hansson E. Neuropharmacological effects of Phoneutria nigriventer venom on astrocytes. Neurochem Int 2016; 96:13-23. [DOI: 10.1016/j.neuint.2016.04.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/07/2016] [Accepted: 04/14/2016] [Indexed: 11/16/2022]
|
8
|
Verkhratsky A, Parpura V. Physiology of Astroglia: Channels, Receptors, Transporters, Ion Signaling and Gliotransmission. ACTA ACUST UNITED AC 2015. [DOI: 10.4199/c00123ed1v01y201501ngl004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
9
|
Garzón M, Duffy AM, Chan J, Lynch MK, Mackie K, Pickel VM. Dopamine D₂ and acetylcholine α7 nicotinic receptors have subcellular distributions favoring mediation of convergent signaling in the mouse ventral tegmental area. Neuroscience 2013; 252:126-43. [PMID: 23954803 DOI: 10.1016/j.neuroscience.2013.08.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 08/06/2013] [Accepted: 08/06/2013] [Indexed: 11/24/2022]
Abstract
Alpha7 nicotinic acetylcholine receptors (α7nAChRs) mediate nicotine-induced burst-firing of dopamine neurons in the ventral tegmental area (VTA), a limbic brain region critically involved in reward and in dopamine D2 receptor (D2R)-related cortical dysfunctions associated with psychosis. The known presence of α7nAChRs and Gi-coupled D2Rs in dopamine neurons of the VTA suggests that these receptors are targeted to at least some of the same neurons in this brain region. To test this hypothesis, we used electron microscopic immunolabeling of antisera against peptide sequences of α7nACh and D2 receptors in the mouse VTA. Dual D2R and α7nAChR labeling was seen in many of the same somata (co-localization over 97%) and dendrites (co-localization over 49%), where immunoreactivity for each of the receptors was localized to endomembranes as well as to non-synaptic or synaptic plasma membranes often near excitatory-type synapses. In comparison with somata and dendrites, many more small axons and axon terminals were separately labeled for each of the receptors. Thus, single-labeled axon terminals were predominant for both α7nAChR (57.9%) and D2R (89.0%). The majority of the immunolabeled axonal profiles contained D2R-immunoreactivity (81.6%) and formed either symmetric or asymmetric synapses consistent with involvement in the release of both inhibitory and excitatory transmitters. Of 160 D2R-labeled terminals, 81.2% were presynaptic to dendrites that expressed α7nAChR alone or together with the D2R. Numerous glial processes inclusive of those enveloping either excitatory- or inhibitory-type synapses also contained single labeling for D2R (n=152) and α7nAChR (n=561). These results suggest that classic antipsychotic drugs, all of which block the D2R, may facilitate α7nAChR-mediated burst-firing by elimination of D2R-dependent inhibition in neurons expressing both receptors as well as by indirect pre-synaptic and glial mechanisms.
Collapse
Affiliation(s)
- M Garzón
- Brain and Mind Research Institute, Weill Cornell Medical College, 407 East 61st Street, New York, NY 10065, USA; Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina UAM, Madrid 28029, Spain; Instituto de Investigación Hospital Universitario La Paz (IDIPAZ), Paseo de la Castellana 261, Madrid 28046, Spain
| | | | | | | | | | | |
Collapse
|
10
|
Modulation of pineal melatonin synthesis by glutamate involves paracrine interactions between pinealocytes and astrocytes through NF-κB activation. BIOMED RESEARCH INTERNATIONAL 2013; 2013:618432. [PMID: 23984387 PMCID: PMC3747608 DOI: 10.1155/2013/618432] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 06/28/2013] [Indexed: 01/05/2023]
Abstract
The glutamatergic modulation of melatonin synthesis is well known, along with the importance of astrocytes in mediating glutamatergic signaling in the central nervous system. Pinealocytes and astrocytes are the main cell types in the pineal gland. The objective of this work was to investigate the interactions between astrocytes and pinealocytes as a part of the glutamate inhibitory effect on melatonin synthesis. Rat pinealocytes isolated or in coculture with astrocytes were incubated with glutamate in the presence of norepinephrine, and the melatonin content, was quantified. The expression of glutamate receptors, the intracellular calcium content and the NF-κB activation were analyzed in astrocytes and pinealocytes. TNF-α's possible mediation of the effect of glutamate was also investigated. The results showed that glutamate's inhibitory effect on melatonin synthesis involves interactions between astrocytes and pinealocytes, possibly through the release of TNF-α. Moreover, the activation of the astrocytic NF-κB seems to be a necessary step. In astrocytes and pinealocytes, AMPA, NMDA, and group I metabotropic glutamate receptors were observed, as well as the intracellular calcium elevation. In conclusion, there is evidence that the modulation of melatonin synthesis by glutamate involves paracrine interactions between pinealocytes and astrocytes through the activation of the astrocytic NF-κB transcription factor and possibly by subsequent TNF-α release.
Collapse
|
11
|
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: 4.0] [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
|
12
|
Molinaro P, Cataldi M, Cuomo O, Viggiano D, Pignataro G, Sirabella R, Secondo A, Boscia F, Pannaccione A, Scorziello A, Sokolow S, Herchuelz A, Di Renzo G, Annunziato L. Genetically modified mice as a strategy to unravel the role played by the Na(+)/Ca (2+) exchanger in brain ischemia and in spatial learning and memory deficits. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 961:213-22. [PMID: 23224882 DOI: 10.1007/978-1-4614-4756-6_18] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Because no isoform-specific blocker of NCX has ever been synthesized, a more selective strategy to identify the role of each antiporter isoform in the brain was represented by the generation of knockout and knockin mice for the different isoforms of the antiporter.Experiments performed in NCX2 and NCX3 knockout mice provided evidence that these two isoforms participate in spatial learning and memory consolidation, although in an opposite manner. These new data from ncx2-/- and ncx3-/- mice may open new experimental avenues for the development of effective therapeutic compounds that, by selectively inhibiting or activating these molecular targets, could treat patients affected by cognitive impairment including Alzheimer's, Parkinson's, Huntington's diseases, and infarct dementia.More importantly, knockout and knockin mice also provided new relevant information on the role played by NCX in maintaining the intracellular Na(+) and Ca(2+) homeostasis and in protecting neurons during brain ischemia. In particular, both ncx2-/- and ncx3-/- mice showed an increased neuronal vulnerability after the ischemic insult induced by transient middle cerebral artery occlusion.As the ubiquitous deletion of NCX1 brings about to an early death of embryos because of a lack of heartbeat, this strategy could not be successfully pursued. However, information on the role of NCX1 in normal and ischemic brain could be obtained by developing conditional knockout mice lacking NCX1 in the brain. Preliminarily results obtained in these conditional mice suggest that also NCX1 protects neurons from ischemic cell death.Overall, the use of genetic-modified mice for NCX1, NCX2, and NCX3 represents a fruitful strategy to characterize the physiological role exerted by NCX in CNS and to identify the isoforms of the antiporter as potential molecular targets for therapeutic intervention in cerebral ischemia.
Collapse
Affiliation(s)
- Pasquale Molinaro
- Department of Neuroscience, Federico II University of Naples, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Thyssen A, Stavermann M, Buddrus K, Doengi M, Ekberg JA, St John JA, Deitmer JW, Lohr C. Spatial and developmental heterogeneity of calcium signaling in olfactory ensheathing cells. Glia 2012; 61:327-37. [PMID: 23109369 DOI: 10.1002/glia.22434] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 09/11/2012] [Indexed: 12/24/2022]
Abstract
Olfactory ensheathing cells (OECs) are specialized glial cells in the mammalian olfactory system supporting growth of axons from the olfactory epithelium into the olfactory bulb. OECs in the olfactory bulb can be subdivided into OECs of the outer nerve layer and the inner nerve layer according to the expression of marker proteins and their location in the nerve layer. In the present study, we have used confocal calcium imaging of OECs in acute mouse brain slices and olfactory bulbs in toto to investigate physiological differences between OEC subpopulations. OECs in the outer nerve layer, but not the inner nerve layer, responded to glutamate, ATP, serotonin, dopamine, carbachol, and phenylephrine with increases in the cytosolic calcium concentration. The calcium responses consisted of a transient and a tonic component, the latter being mediated by store-operated calcium entry. Calcium measurements in OECs during the first three postnatal weeks revealed a downregulation of mGluR(1) and P2Y(1) receptor-mediated calcium signaling within the first 2 weeks, suggesting that the expression of these receptors is developmentally controlled. In addition, electrical stimulation of sensory axons evoked calcium signaling via mGluR(1) and P2Y(1) only in outer nerve layer OECs. Downregulation of the receptor-mediated calcium responses in postnatal animals is reflected by a decrease in amplitude of stimulation-evoked calcium transients in OECs from postnatal days 3 to 21. In summary, the results presented reveal striking differences in receptor responses during development and in axon-OEC communication between the two subpopulations of OECs in the olfactory bulb.
Collapse
Affiliation(s)
- Anne Thyssen
- Abteilung für Allgemeine Zoologie, TU Kaiserslautern, Germany
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Verkhratsky A, Rodríguez JJ, Parpura V. Calcium signalling in astroglia. Mol Cell Endocrinol 2012; 353:45-56. [PMID: 21945602 DOI: 10.1016/j.mce.2011.08.039] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 08/28/2011] [Accepted: 08/31/2011] [Indexed: 12/15/2022]
Abstract
Astroglia possess excitability based on movements of Ca(2+) ions between intracellular compartments and plasmalemmal Ca(2+) fluxes. This "Ca(2+) excitability" is controlled by several families of proteins located in the plasma membrane, within the cytosol and in the intracellular organelles, most notably in the endoplasmic reticulum (ER) and mitochondria. Accumulation of cytosolic Ca(2+) can be caused by the entry of Ca(2+) from the extracellular space through ionotropic receptors and store-operated channels expressed in astrocytes. Plasmalemmal Ca(2+) ATP-ase and sodium-calcium exchanger extrude cytosolic Ca(2+) to the extracellular space; the exchanger can also operate in reverse, depending of the intercellular Na(+) concentration, to deliver Ca(2+) to the cytosol. The ER internal store possesses inositol 1,4,5-trisphosphate receptors which can be activated upon stimulation of astrocytes through a multiple plasma membrane metabotropic G-protein coupled receptors. This leads to release of Ca(2+) from the ER and its elevation in the cytosol, the level of which can be modulated by mitochondria. The mitochondrial uniporter takes up Ca(2+) into the matrix, while free Ca(2+) exits the matrix through the mitochondrial Na(+)/Ca(2+) exchanger as well as via transient openings of the mitochondrial permeability transition pore. One of the prominent consequences of astroglial Ca(2+) excitability is gliotransmission, a release of transmitters from astroglia which can lead to signalling to adjacent neurones.
Collapse
|
15
|
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
|
16
|
Ahmed Z, Wieraszko A. Trans-spinal direct current enhances corticospinal output and stimulation-evoked release of glutamate analog, D-2,3-³H-aspartic acid. J Appl Physiol (1985) 2012; 112:1576-92. [PMID: 22362399 DOI: 10.1152/japplphysiol.00967.2011] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Trans-spinal direct current (tsDC) stimulation is a modulator of spinal excitability and can influence cortically elicited muscle contraction in a polarity-dependent fashion. When combined with low-frequency repetitive cortical stimulation, cathodal tsDC [tsDC(-)] produces a long-term facilitation of cortically elicited muscle actions. We investigated the ability of this combined stimulation paradigm to facilitate cortically elicited muscle actions in spinal cord-injured and noninjured animals. The effect of tsDC-applied alone or in combination with repetitive spinal stimulation (rSS) on the release of the glutamate analog, D-2,3-(3)H-aspartate (D-Asp), from spinal cord preparations in vitro-was also tested. In noninjured animals, tsDC (-2 mA) reproducibly potentiated cortically elicited contractions of contralateral and ipsilateral muscles tested at various levels of baseline muscle contraction forces. Cortically elicited muscle responses in animals with contusive and hemisectioned spinal cord injuries (SCIs) were similarly potentiated. The combined paradigm of stimulation caused long-lasting potentiation of cortically elicited bilateral muscle contraction in injured and noninjured animals. Additional analysis suggests that at higher baseline forces, tsDC(-) application does not increase the rising slope of the muscle contraction but causes repeated firing of the same motor units. Both cathodal and anodal stimulations induced a significant increase of D-Asp release in vitro. The effect of the combined paradigm of stimulation (tsDC and rSS) on the concentration of extracellular D-Asp was polarity dependent. These results indicate that tsDC can powerfully modulate the responsiveness of spinal cord neurons. The results obtained from the in vitro preparation suggest that the changes in neuronal excitability were correlated with an increased concentration of extracellular glutamate. The combined paradigm of stimulation, used in our experiments, could be noninvasively applied to restore motor control in humans with SCI.
Collapse
Affiliation(s)
- Zaghloul Ahmed
- Dept. of Physical Therapy, College of Staten Island, 2800 Victory Blvd., Rm. 202, Staten Island, NY 10314, USA.
| | | |
Collapse
|
17
|
Abstract
The name astroglia unifies many non-excitable neural cells that act as primary homeostatic cells in the nervous system. Neuronal activity triggers multiple homeostatic responses of astroglia that include increase in metabolic activity and synthesis of neuronal preferred energy substrate lactate, clearance of neurotransmitters and buffering of extracellular K(+) ions to name but a few. Many (if not all) of astroglial homeostatic responses are controlled by dynamic changes in the cytoplasmic concentration of two cations, Ca(2+) and Na(+). Intracellular concentration of these ions is tightly controlled by several transporters and can be rapidly affected by the activation of respective fluxes through ionic channels or ion exchangers. Here, we provide a comprehensive review of astroglial Ca(2+) and Na(+) signalling.
Collapse
|
18
|
Duffy AM, Fitzgerald ML, Chan J, Robinson DC, Milner TA, Mackie K, Pickel VM. Acetylcholine α7 nicotinic and dopamine D2 receptors are targeted to many of the same postsynaptic dendrites and astrocytes in the rodent prefrontal cortex. Synapse 2011; 65:1350-67. [PMID: 21858872 PMCID: PMC3356922 DOI: 10.1002/syn.20977] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The alpha-7 nicotinic acetylcholine receptor (α7nAChR) and the dopamine D(2) receptor (D(2) R) are both implicated in attentional processes and cognition, mediated in part through the prefrontal cortex (PFC). We examined the dual electron microscopic immunolabeling of α7nAChR and either D(2) R or the vesicular acetylcholine transporter (VAChT) in rodent PFC to assess convergent functional activation sites. Immunoreactivity (ir) for α7nAChR and/or D(2) R was seen in the same as well as separate neuronal and glial profiles. At least half of the dually labeled profiles were somata and dendrites, while most labeled axon terminals expressed only D(2) R-ir. The D(2) R-labeled terminals were without synaptic specializations or formed inhibitory or excitatory-type synapses with somatodendritic profiles, some of which expressed the α7nAChR and/or D(2) R. Astrocytic glial processes comprised the majority of nonsomatodendritic α7nAChR or α7nAChR and D(2) R-labeled profiles. Glial processes containing α7nAChR-ir were frequently located near VAChT-labeled terminals and also showed perisynaptic and perivascular associations. We conclude that in rodent PFC α7nACh and D(2) R activation can dually modulate (1) postsynaptic dendritic responses within the same or separate but synaptically linked neurons in which the D(2) R has the predominately presynaptic distribution, and (2) astrocytic signaling that may be crucial for synaptic transmission and functional hyperemia.
Collapse
Affiliation(s)
- Aine M. Duffy
- Department of Neurology and Neuroscience, Division of Neurobiology, Weill Cornell Medical College, New York, New York 10065
| | - Megan L. Fitzgerald
- Department of Neurology and Neuroscience, Division of Neurobiology, Weill Cornell Medical College, New York, New York 10065
| | - June Chan
- Department of Neurology and Neuroscience, Division of Neurobiology, Weill Cornell Medical College, New York, New York 10065
| | - Danielle C. Robinson
- Department of Neurology and Neuroscience, Division of Neurobiology, Weill Cornell Medical College, New York, New York 10065
| | - Teresa A. Milner
- Department of Neurology and Neuroscience, Division of Neurobiology, Weill Cornell Medical College, New York, New York 10065
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York 10065
| | - Kenneth Mackie
- Department of Psychological and Brain Sciences and the Gill Center, Indiana University, Bloomington, Indiana 47405
| | - Virginia M. Pickel
- Department of Neurology and Neuroscience, Division of Neurobiology, Weill Cornell Medical College, New York, New York 10065
| |
Collapse
|
19
|
Abstract
Astrocytes were identified about 150 years ago, and, for the longest time, were considered to be supporting cells in the brain providing trophic, metabolic, and structural support for neural networks. Research in the last 2 decades has uncovered many novel molecules in astrocytes and the finding that astrocytes communicate with neurons via Ca2+ signaling, which leads to release of chemical transmitters, termed gliotransmitters, has led to renewed interest in their biology. This chapter will briefly review the unique morphology and molecular properties of astrocytes. The reader will be introduced to the role of astrocytes in blood-brain barrier (BBB) maintenance, in Ca2+ signaling, in synaptic transmission, in CNS synaptogenesis, and as neural progenitor cells. Mention is also made of the diseases in which astrocyte dysfunction has a role.
Collapse
Affiliation(s)
- Sukriti Nag
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
20
|
Prebil M, Vardjan N, Jensen J, Zorec R, Kreft M. Dynamic monitoring of cytosolic glucose in single astrocytes. Glia 2011; 59:903-13. [PMID: 21381116 DOI: 10.1002/glia.21161] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Accepted: 01/25/2011] [Indexed: 12/31/2022]
Abstract
It is becoming increasingly clear that astrocytes are no longer playing a subservient role to neurons in the central nervous system (CNS), and that these cells are being considered as active communication integrators. They respond to neurotransmitters by the regulated release of gliotransmitters. The delay between neurotransmitter activation and the release of gliotransmitters from astrocytes is in the time-domain of subseconds, much slower than the submillisecond synaptic delay. Astrocytes also control microcirculation and provide metabolic support for neurons. However, the dynamics of their energy metabolic response to neurotransmitter application is not known. We here used a FRET glucose nanosensor to dynamically measure the cytosolic glucose concentration in single astrocytes. We show that following the adrenaline or noradrenaline stimulation the availability of cytosolic glucose is increased promptly after stimulation with a time-constant of 116.7 s and 115.9 s, respectively. A decline in cytosolic glucose concentration with a time-constant of 50.7 s was observed during glutamate and 16.7 s during lactate addition to astrocytes, when these were bathed in the presence of extracellular glucose-containing solution, likely reflecting predominant glucose engagement in glycogen synthesis. In contrast, in the glucose-free extracellular solution, glutamate application to astrocytes resulted in a slow increase in cytosolic glucose concentration, consistent with the view that glutamate may be an alternative energy source in hypoglycemic conditions. We conclude that astrocytic cytosolic glucose metabolism responds in the time-domain of tens of seconds, which is slower compared to the whole brain functional magnetic resonance imaging measurements of the local intravascular hemodynamic response.
Collapse
Affiliation(s)
- Mateja Prebil
- Faculty of Medicine, Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Zaloska 4, Ljubljana, Slovenia
| | | | | | | | | |
Collapse
|
21
|
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: 9.5] [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
|
22
|
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: 7.8] [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
|
23
|
Nonlinear gap junctions enable long-distance propagation of pulsating calcium waves in astrocyte networks. PLoS Comput Biol 2010; 6. [PMID: 20865153 PMCID: PMC2928752 DOI: 10.1371/journal.pcbi.1000909] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Accepted: 07/28/2010] [Indexed: 11/28/2022] Open
Abstract
A new paradigm has recently emerged in brain science whereby communications between glial cells and neuron-glia interactions should be considered together with neurons and their networks to understand higher brain functions. In particular, astrocytes, the main type of glial cells in the cortex, have been shown to communicate with neurons and with each other. They are thought to form a gap-junction-coupled syncytium supporting cell-cell communication via propagating Ca2+ waves. An identified mode of propagation is based on cytoplasm-to-cytoplasm transport of inositol trisphosphate (IP3) through gap junctions that locally trigger Ca2+ pulses via IP3-dependent Ca2+-induced Ca2+ release. It is, however, currently unknown whether this intracellular route is able to support the propagation of long-distance regenerative Ca2+ waves or is restricted to short-distance signaling. Furthermore, the influence of the intracellular signaling dynamics on intercellular propagation remains to be understood. In this work, we propose a model of the gap-junctional route for intercellular Ca2+ wave propagation in astrocytes. Our model yields two major predictions. First, we show that long-distance regenerative signaling requires nonlinear coupling in the gap junctions. Second, we show that even with nonlinear gap junctions, long-distance regenerative signaling is favored when the internal Ca2+ dynamics implements frequency modulation-encoding oscillations with pulsating dynamics, while amplitude modulation-encoding dynamics tends to restrict the propagation range. As a result, spatially heterogeneous molecular properties and/or weak couplings are shown to give rise to rich spatiotemporal dynamics that support complex propagation behaviors. These results shed new light on the mechanisms implicated in the propagation of Ca2+ waves across astrocytes and the precise conditions under which glial cells may participate in information processing in the brain. In recent years, the focus of Cellular Neuroscience has progressively stopped only being on neurons but started to include glial cells as well. Indeed, astrocytes, the main type of glial cells in the cortex, dynamically modulate neuron excitability and control the flow of information across synapses. Moreover, astrocytes have been shown to communicate with each other over long distances using calcium waves. These waves spread from cell to cell via molecular gates called gap junctions, which connect neighboring astrocytes. In this work, we used a computer model to question what biophysical mechanisms could support long-distance propagation of Ca2+ wave signaling. The model shows that the coupling function of the gap junction must be non-linear and include a threshold. This prediction is largely unexpected, as gap junctions are classically considered to implement linear functions. Recent experimental observations, however, suggest their operation could actually be more complex, in agreement with our prediction. The model also shows that the distance traveled by waves depends on characteristics of the internal astrocyte dynamics. In particular, long-distance propagation is facilitated when internal calcium oscillations are in their frequency-modulation encoding mode and are pulsating. Hence, this work provides testable experimental predictions to decipher long-distance communication between astrocytes.
Collapse
|
24
|
Hoffmann A, Grimm C, Kraft R, Goldbaum O, Wrede A, Nolte C, Hanisch UK, Richter-Landsberg C, Brück W, Kettenmann H, Harteneck C. TRPM3 is expressed in sphingosine-responsive myelinating oligodendrocytes. J Neurochem 2010; 114:654-65. [PMID: 20163522 DOI: 10.1111/j.1471-4159.2010.06644.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Oligodendrocytes are the myelin-forming cells of the CNS and guarantee proper nerve conduction. Sphingosine, one major component of myelin, has recently been identified to activate TRPM3, a member of the melastatin-related subfamily of transient receptor potential (TRP) channels. TRPM3 has been demonstrated to be expressed in brain with unknown cellular distribution. Here, we show for the first time that TRPM3 is expressed in oligodendrocytes in vitro and in vivo. TRPM3 is present during oligodendrocyte differentiation. Immunohistochemistry of adult rat brain slices revealed staining of white matter areas, which co-localized with oligodendrocyte markers. Analysis of the developmental distribution revealed that, prior to myelination, TRPM3 channels are localized on neurons. On oligodendrocytes they are found after the onset of myelination. RT-PCR studies showed that the transcription of TRPM3 splice variants is also developmentally regulated in vitro. Ca(2+) imaging approaches revealed the presence of a sphingosine-induced Ca(2+) entry mechanism in oligodendrocytes - with a pharmacological profile similar to the profile published for heterologously expressed TRPM3. These findings indicate that TRPM3 participates as a Ca(2+)-permeable and sphingosine-activated channel in oligodendrocyte differentiation and CNS myelination.
Collapse
Affiliation(s)
- Anja Hoffmann
- Zelluläre Neurowissenschaften, Max-Delbrück-Centrum, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Mandal A, Shahidullah M, Delamere NA. Hydrostatic pressure-induced release of stored calcium in cultured rat optic nerve head astrocytes. Invest Ophthalmol Vis Sci 2010; 51:3129-38. [PMID: 20071675 DOI: 10.1167/iovs.09-4614] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Elevated intraocular pressure is associated with glaucomatous optic nerve damage. Other investigators have shown functional changes in optic nerve head astrocytes subjected to elevated hydrostatic pressure (HP) for 1 to 5 days. Recently, the authors reported ERK1/2, p90(RSK) and NHE1 phosphorylation after 2 hours. Here they examine calcium responses at the onset of HP to determine what precedes ERK1/2 phosphorylation. METHODS Cytoplasmic calcium concentration ([Ca(2+)](i)) was measured in cultured rat optic nerve astrocytes loaded with fura-2. The cells were placed in a closed imaging chamber and subjected to an HP increase of 15 mm Hg. Protein phosphorylation was detected by Western blot analysis. RESULTS The increase of HP caused an immediate slow increase in [Ca(2+)](i). The response persisted in calcium-free solution and when nickel chloride (4 mM) was added to suppress channel-mediated calcium entry. Previous depletion of the ER calcium stores by cyclopiazonic acid abolished the HP-induced calcium level increase. The HP-induced increase persisted in cells exposed to xestospongin C, an inhibitor of IP3R-mediated calcium release. In contrast, ryanodine receptor (RyR) antagonist ruthenium red (10 microM) or dantrolene (25 microM) inhibited the HP-induced calcium increase. The HP-induced calcium increase was abolished when ryanodine-sensitive calcium stores were pre-depleted with caffeine (3 mM). HP caused ERK1/2 phosphorylation. The magnitude of the ERK1/2 phosphorylation response was reduced by ruthenium red and dantrolene. CONCLUSIONS Increasing HP causes calcium release from a ryanodine-sensitive cytoplasmic store and subsequent ERK1/2 activation. Calcium store release appears to be a required early step in the initial astrocyte response to an HP increase.
Collapse
Affiliation(s)
- Amritlal Mandal
- Department of Physiology, University of Arizona, Tucson, Arizona 85724, USA
| | | | | |
Collapse
|
26
|
Kawai T, Abe H, Wakabayashi KI, Oka Y. Calcium oscillations in the olfactory nonsensory cells of the goldfish, Carassius auratus. Biochim Biophys Acta Gen Subj 2009; 1790:1681-8. [PMID: 19800938 DOI: 10.1016/j.bbagen.2009.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 09/02/2009] [Accepted: 09/28/2009] [Indexed: 11/17/2022]
Abstract
BACKGROUND The olfactory nonsensory cells contribute to the maintenance of normal functions of the olfactory epithelium (OE). Specifically, the ciliated nonsensory cells of teleosts play important roles in the odorant detection by OE in aqueous environment. Their cilia show strong beating activities and cause water flow at the OE surface, making the detection of odorants by OE more efficient. Because intracellular Ca2+ level has been reported to play an important role in ciliary beating, the ciliary beating activity may be regulated by intracellular Ca2+ dynamics of these ciliated nonsensory cells. METHODS We performed Ca2+ imaging experiments to analyze the Ca2+ dynamics in acutely dissociated OE cells of the goldfish. Furthermore, we examined the contribution of the Ca2+ dynamics to the ciliary beating frequency (CBF) at the surface of the intact OE. RESULTS Olfactory nonsensory cells showed both spontaneous intracellular Ca2+ oscillations and propagating intercellular Ca2+ waves. Application of 2-aminoethoxydiphenylborate (2-APB), which antagonizes IP3-induced Ca2+ release from intracellular stores suppressed these Ca2+ oscillations. Furthermore, 2-APB application to the intact OE lamellae resulted in the decrease of CBF at the surface of the OE. CONCLUSIONS These results indicate that spontaneous intracellular calcium oscillations persistently up-regulate the ciliary beating at the surface of the OE in teleosts. GENERAL SIGNIFICANCE Ciliary beating activity at the surface of OE can be regulated by the Ca2+ dynamics of olfactory nonsensory cells. Because this ciliary movement causes inflow of external fluid into the nostril, this regulation is suggested to influence the efficiency of odorant detection by OE.
Collapse
Affiliation(s)
- Takafumi Kawai
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | |
Collapse
|
27
|
OU J, KUMAR Y, ALIOUA A, SAILER C, STEFANI E, TORO L. Ca2+- and thromboxane-dependent distribution of MaxiK channels in cultured astrocytes: from microtubules to the plasma membrane. Glia 2009; 57:1280-95. [PMID: 19170178 PMCID: PMC2713352 DOI: 10.1002/glia.20847] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Large-conductance, voltage- and Ca2+-activated K+ channels (MaxiK) are broadly expressed ion channels minimally assembled by four pore-forming alpha-subunits (MaxiKalpha) and typically observed as plasma membrane proteins in various cell types. In murine astrocyte primary cultures, we show that MaxiKalpha is predominantly confined to the microtubule network. Distinct microtubule distribution of MaxiKalpha was visualized by three independent labeling approaches: (1) MaxiKalpha-specific antibodies, (2) expressed EGFP-labeled MaxiKalpha, and (3) fluorophore-conjugated iberiotoxin, a specific MaxiK pore-blocker. This MaxiKalpha association with microtubules was further confirmed by in vitro His-tag pulldown, co-immunoprecipitation from brain lysates, and microtubule depolymerization experiments. Changes in intracellular Ca2+ elicited by general pharmacological agents, caffeine or thapsigargin, resulted in increased MaxiKalpha labeling at the plasma membrane. More notably, U46619, an analog of thromboxane A2 (TXA2), which triggers Ca2+-release pathways and whose levels increase during cerebral hemorrhage/trauma, also elicits a similar increase in MaxiKalpha surface labeling. Whole-cell patch clamp recordings of U46619-stimulated cells develop a approximately 3-fold increase in current amplitude indicating that TXA2 stimulation results in the recruitment of additional, functional MaxiK channels to the surface membrane. While microtubules are largely absent in mature astrocytes, immunohistochemistry results in brain slices show that cortical astrocytes in the newborn mouse (P1) exhibit a robust expression of microtubules that significantly colocalize with MaxiK. The results of this study provide the novel insight that suggests that Ca2+ released from intracellular stores may play a key role in regulating the traffic of intracellular, microtubule-associated MaxiK stores to the plasma membrane of developing murine astrocytes.
Collapse
Affiliation(s)
- J.W. OU
- Department of Molecular and Medical Pharmacology, University of California at Los Angeles, Los Angeles, CA 90095
| | - Y. KUMAR
- Department of Anesthesiology, University of California at Los Angeles, Los Angeles, CA 90095
| | - A. ALIOUA
- Department of Anesthesiology, University of California at Los Angeles, Los Angeles, CA 90095
| | - C. SAILER
- Division for Molecular and Cellular Pharmacology, Medical University, Innsbruck, Peter Mayr-Strasse 1, A-6020 Innsbruck, Austria
| | - E. STEFANI
- Department of Anesthesiology, University of California at Los Angeles, Los Angeles, CA 90095
- Department of Physiology, University of California at Los Angeles, Los Angeles, CA 90095
- Brain Research Institute, University of California at Los Angeles, Los Angeles, CA 90095
| | - L. TORO
- Department of Molecular and Medical Pharmacology, University of California at Los Angeles, Los Angeles, CA 90095
- Department of Anesthesiology, University of California at Los Angeles, Los Angeles, CA 90095
- Brain Research Institute, University of California at Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
28
|
Glutamate regulation of calcium and IP3 oscillating and pulsating dynamics in astrocytes. J Biol Phys 2009; 35:383-411. [PMID: 19669422 DOI: 10.1007/s10867-009-9155-y] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2009] [Accepted: 04/14/2009] [Indexed: 10/20/2022] Open
Abstract
Recent years have witnessed an increasing interest in neuron-glia communication. This interest stems from the realization that glia participate in cognitive functions and information processing and are involved in many brain disorders and neurodegenerative diseases. An important process in neuron-glia communications is astrocyte encoding of synaptic information transfer-the modulation of intracellular calcium (Ca(2+)) dynamics in astrocytes in response to synaptic activity. Here, we derive and investigate a concise mathematical model for glutamate-induced astrocytic intracellular Ca(2+) dynamics that captures the essential biochemical features of the regulatory pathway of inositol 1,4,5-trisphosphate (IP(3)). Starting from the well-known two-variable (intracellular Ca(2+) and inactive IP(3) receptors) Li-Rinzel model for calcium-induced calcium release, we incorporate the regulation of IP(3) production and phosphorylation. Doing so, we extend it to a three-variable model (which we refer to as the ChI model) that could account for Ca(2+) oscillations with endogenous IP(3) metabolism. This ChI model is then further extended into the G-ChI model to include regulation of IP(3) production by external glutamate signals. Compared with previous similar models, our three-variable models include a more realistic description of IP(3) production and degradation pathways, lumping together their essential nonlinearities within a concise formulation. Using bifurcation analysis and time simulations, we demonstrate the existence of new putative dynamical features. The cross-couplings between IP(3) and Ca(2+) pathways endow the system with self-consistent oscillatory properties and favor mixed frequency-amplitude encoding modes over pure amplitude-modulation ones. These and additional results of our model are in general agreement with available experimental data and may have important implications for the role of astrocytes in the synaptic transfer of information.
Collapse
|
29
|
Di Garbo A. Dynamics of a minimal neural model consisting of an astrocyte, a neuron, and an interneuron. J Biol Phys 2009; 35:361-82. [PMID: 19669428 DOI: 10.1007/s10867-009-9143-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Accepted: 02/16/2009] [Indexed: 11/26/2022] Open
Abstract
In this paper, a biophysical neural network model consisting of a pyramidal neuron, an interneuron, and the astrocyte is studied. The corresponding dynamical properties are mainly investigated by using numerical simulations. The results show that the presence of the adenosine triphosphate and of the interneuron impacts the overall neural activity. It is shown that the fluxes of calcium through the cellular membrane strongly affect the modulation of the neural activity arising from the astrocyte.
Collapse
|
30
|
De Pittà M, Volman V, Levine H, Ben-Jacob E. Multimodal encoding in a simplified model of intracellular calcium signaling. Cogn Process 2008; 10 Suppl 1:S55-70. [DOI: 10.1007/s10339-008-0242-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2008] [Revised: 10/26/2008] [Accepted: 10/31/2008] [Indexed: 11/24/2022]
|
31
|
Takahashi-Iwanaga H, Nio-Kobayashi J, Habara Y, Furuya K. A dual system of intercellular calcium signaling in glial nets associated with lanceolate sensory endings in rat vibrissae. J Comp Neurol 2008; 510:68-78. [PMID: 18615537 DOI: 10.1002/cne.21756] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The lanceolate sensory endings that form palisades around the hair follicle associate with networks of branched Schwann cells. To define the properties of these glial networks as possible conduits of Ca2+ signals, lanceolate endings isolated from rat vibrissae were observed by confocal microscopy while the signaling was locally activated by mechanical stimulation. Intercellular coupling by gap junctions was also assessed by a technique employing fluorescence recovery after photobleaching (FRAP) and by transmission electron microscopy (TEM). Results showed that the glial Ca2+ signals can spread among the arrays of lanceolates in two forms: rapid signals that originate in individual Schwann processes covering the lanceolate axon terminals around the locus of mechanical stimulation, and delayed ones that travel from the stimulation locus through cytoplasmic arborization of the primarily activated cell to the adjacent cell processes. The former signaling was suppressed by the antipurinergic agents suramin and apyrase, whereas the latter was sensitive to the gap junction blocker carbenoxolon. FRAP experiments and TEM observations corroborated the presence of gap junction communications between the Schwann processes of different cell origins. These findings show that, in the Schwann networks, purinergically induced Ca2+ signals and those dependent on gap junctions are propagated in their own spatiotemporal patterns to constitute two distinct forms of communication among the mechanoreceptor palisades.
Collapse
Affiliation(s)
- Hiromi Takahashi-Iwanaga
- Laboratory of Histology and Cytology, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan.
| | | | | | | |
Collapse
|
32
|
|
33
|
Swiatek-De Lange M, Stampfl A, Hauck SM, Zischka H, Gloeckner CJ, Deeg CA, Ueffing M. Membrane-initiated effects of progesterone on calcium dependent signaling and activation of VEGF gene expression in retinal glial cells. Glia 2007; 55:1061-73. [PMID: 17551930 DOI: 10.1002/glia.20523] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Neurosteroids, such as progesterone, influence central nervous system development and function by regulating a broad spectrum of physiological processes. Here, we investigated membrane-initiated actions of progesterone in the retina and identified the membrane-associated progesterone receptor component 1 (PGRMC1). We found PGRMC1 expressed mainly in retinal Muller glia (RMG) and retinal pigment epithelium, and localized uniquely to microsomal and plasma membrane fractions. In RMG, membrane-impermeable progesterone conjugate induced calcium influx and subsequent phosphatidylinositol 3-kinase-mediated phosphorylation of PKC and ERK-1/2. Induction by progesterone also led to PKC-dependent activation of VEGF gene expression and protein synthesis, suggesting a contribution of membrane-initiated hormone effects to VEGF induced neovascularization within retina.
Collapse
Affiliation(s)
- Magdalena Swiatek-De Lange
- Institute of Human Genetics, GSF National-Research Center for Environment and Health, Neuherberg, Germany
| | | | | | | | | | | | | |
Collapse
|
34
|
Lee Y, Gaskins D, Anand A, Shekhar A. Glia mechanisms in mood regulation: a novel model of mood disorders. Psychopharmacology (Berl) 2007; 191:55-65. [PMID: 17225169 DOI: 10.1007/s00213-006-0652-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2006] [Accepted: 11/14/2006] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Recent evidence in clinical and preclinical studies has implicated glutamate neurotransmissions in pathophysiology of mood disorders. The regulation of amino acid neurotransmission, i.e., glutamate and gamma-aminobutyric acid (GABA) involves coordinated mechanisms of uptake and transport within a tripartite synaptic system that includes neurons and glia. Newly appreciated role of the glia, more specifically astrocytes on neuronal functions combined with reported postmortem abnormalities of glia in patients with mood disorders further supports the role of glia in mood disorders. MATERIALS AND METHODS This report presents some of our preliminary results utilizing glia-selective toxins and other pharmacological tools to suppress glial function within the limbic system to study the resulting behavioral abnormalities, and thus, elucidate glial involvement in the development of mood disorders. RESULTS AND DISCUSSION We demonstrate that chronic blockade of glutamate uptake by a glial/neuronal transporter antagonist L-trans-pyrrolidine-2,4-dicarboxylic acid (PDC) within the amygdala, a key area implicated in mood regulation, results in dose-dependent reduction in social exploratory behavior and disrupts circadian activity patterns consistent with symptoms of mood disorders. Similarly, the selective astrocytic glutamate transporter type 1 (GLT-1) blocker dihydrokainic acid (DHK) injected into the amygdala also results in reduced social interaction that is blocked by selective glutamate N-methyl-D-aspartate (NMDA) type receptor antagonist AP5. The results are discussed in the context of glial and glutamate mechanisms in mood disorders and potential therapeutic avenues to address these mechanisms.
Collapse
Affiliation(s)
- Younglim Lee
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | |
Collapse
|
35
|
Di Garbo A, Barbi M, Chillemi S, Alloisio S, Nobile M. Calcium signalling in astrocytes and modulation of neural activity. Biosystems 2006; 89:74-83. [PMID: 17196325 DOI: 10.1016/j.biosystems.2006.05.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2005] [Accepted: 05/24/2006] [Indexed: 11/30/2022]
Abstract
Starting from the experimental data on ATP evoked calcium responses in astrocytes, a biophysical model describing these phenomena was built. The simulations showed, in agreement with the experimental findings, that the intracellular calcium fluxes mediated by the P2X and P2Y purinoreceptors are responsible for the biphasic ATP evoked calcium response in astrocytes. Then, the modulation effects on the neural dynamics arising from the release of glutamate from astrocyte are also investigated. By using a minimal network model describing a neuron coupled to the astrocyte, we demonstrated that the calcium extrusion rate through the astrocyte membrane is critically involved in the generation of different firing patterns of the neuron.
Collapse
Affiliation(s)
- Angelo Di Garbo
- Istituto di Biofisica CNR, Via G. Moruzzi 1, 56124 Pisa, Italy.
| | | | | | | | | |
Collapse
|
36
|
Verkhratsky A. Patching the glia reveals the functional organisation of the brain. Pflugers Arch 2006; 453:411-20. [PMID: 16775706 DOI: 10.1007/s00424-006-0099-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2006] [Accepted: 04/28/2006] [Indexed: 10/24/2022]
Abstract
The neuroglia was initially conceived by Rudolf Virchow as a non-cellular connective tissue holding neurones together. In 1894, Carl Ludwig Schleich proposed a hypothesis of fully integrated and interconnected neuronal-glial circuits as a substrate for brain function. This hypothesis received direct experimental support only hundred years later, after several physiological techniques, and most notably the patch-clamp method, were applied to glial cells. These experiments have demonstrated the existence of active and bi-directional neuronal-glial communications, integrating neuronal networks and glial syncytium into one functional circuit. The data accumulated during last 15 years prompt rethinking of the neuronal doctrine towards more inclusive concept, which regards both neurones and glia as equally responsible for information processing in the brain.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, 1.124 Stopford Building, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
37
|
Abstract
Electrical stimulation of neurons in brain slices evokes increases in cytoplasmic Ca(2+) in neighboring astrocytes. The present study tests whether similar neuron-to-glial signaling occurs in the isolated rat retina in response to light stimulation. Results demonstrate that Müller cells, the principal retinal glial cells, generate transient increases in Ca(2+) under constant illumination. A flickering light stimulus increases the occurrence of these Ca(2+) transients. Antidromic activation of ganglion cell axons also increases the generation of Müller cell Ca(2+) transients. The increases in Ca(2+) transients evoked by light and antidromic stimulation are blocked by the purinergic antagonist suramin and by TTX. The addition of adenosine greatly potentiates the response to light, with light ON evoking large Ca(2+) increases in Müller cells. Suramin, apyrase (an ATP-hydrolyzing enzyme), and TTX substantially reduce the adenosine-potentiated response. NMDA, metabotropic glutamate, GABA(B), and muscarinic receptor antagonists, in contrast, are mainly ineffective in blocking the response. Light-evoked Ca(2+) responses begin in Müller cell processes within the inner plexiform (synaptic) layer of the retina and then spread into cell endfeet at the inner retinal surface. These results represent the first demonstration that Ca(2+) increases in CNS glia can be evoked by a natural stimulus (light flashes). The results suggest that neuron-to-glia signaling in the retina is mediated by neuronal release of ATP, most likely from amacrine and/or ganglion cells, and that the response is augmented under pathological conditions when adenosine levels increase.
Collapse
|
38
|
Abstract
Calcium signaling studies in invertebrate glial cells have been performed mainly in the nervous systems of the medicinal leech (Hirudo medicinalis) and the sphinx moth Manduca sexta. The main advantages of studing glial cells in invertebrate nervous systems are the large size of invertebrate glial cells and their easy accessibility for optical and electrophysiological recordings. Glial cells in both insects and annelids express voltage-gated calcium channels and, in the case of leech glial cells, calcium-permeable neurotransmitter receptors, which allow calcium influx as one major source for cytosolic calcium transients. Calcium release from intracellular stores can be induced by metabotropic receptor activation in leech glial cells, but appears to play a minor role in calcium signaling. In glial cells of the antennal lobe of Manduca, voltage-gated calcium signaling changes during postembryonic development and is essential for the migration of the glial cells, a key step in axon guidance and in stabilization of the glomerular structures that are characteristic of primary olfactory centers.
Collapse
Affiliation(s)
- Christian Lohr
- Abteilung für Allgemeine Zoologie, FB Biologie, TU Kaiserslautern, Postfach 3049, 67653 Kaiserslautern, Germany
| | - Joachim W Deitmer
- Abteilung für Allgemeine Zoologie, FB Biologie, TU Kaiserslautern, Postfach 3049, 67653 Kaiserslautern, Germany
| |
Collapse
|
39
|
Abstract
Classic studies have recognized neurons and three glial elements in the central nervous system (CNS) - astrocytes, oligodendrocytes and microglia. The identification of novel glia that specifically express the NG2 chondroitin sulphate proteoglycan (CSPG) raises the possibility of a fifth element. Until recently, all NG2-expressing glia were considered to be oligodendrocyte precursor cells (OPCs) that persist in the adult CNS to generate oligodendrocytes throughout life. However, this narrow view of the function of 'NG2-glia' is being challenged. The majority of NG2-expressing glia in the adult CNS are a distinct class of cells that we have called 'synantocytes' (from the Greek synanto for contact). Synantocytes are stellate cells, with large process arborizations, and are exquisitely related to neurons. Individual cells traverse white and grey matter and form multiple contacts with neurons, astrocytes, oligodendrocytes and myelin. Synantocytes are an integral component of the 'tetrapartite' synapse, and provide a potential integrative neuron-glial communications pathway. Neuronal activity, glutamate and adenosine triphosphate (ATP) act on synantocyte receptors and evoke raised intracellular calcium. It remains to be seen whether this serves a physiological function, but synantocytes may be specialized to monitor signals from neurons and glia, and to respond to changes in the integrity of the CNS via their specific contacts and ion channel and receptor profiles. The general consequences of synantocyte activation are proliferation and phenotypic changes, resulting in glial scar formation, or regeneration of oligodendrocytes, and possibly neurons.
Collapse
Affiliation(s)
- Arthur M Butt
- Wolfson Centre for Age Related Diseases, King's College, London, UK.
| | | | | | | | | |
Collapse
|
40
|
Slamon ND, Mead C, Morgan C, Mitchell A, Pentreath VW. The involvement of calcium in the protective and toxic (nonlinear) responses of rodent and human astroglial cells. NONLINEARITY IN BIOLOGY, TOXICOLOGY, MEDICINE 2005; 3:79-95. [PMID: 19330156 PMCID: PMC2657843 DOI: 10.2201/nonlin.003.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The involvement of [Ca(2+)](i) in the reactive changes of astrocytes which accompany exposure to different chemicals were studied in cultures of C6 and 1321N1 cells. Cells were exposed to up to three serial pulses of the differentiating agent dBcAMP, which induces activation-type changes in the cells. Other cells, with or without the dBcAMP treatments, were treated with a range of concentrations of the antidepressants amitriptyline and fluoxetine and the glial toxicants acrylamide and chloroquine. In some experiments the L-type voltage calcium channel blocker Nifedipine was employed. [Ca(2+)](i) was measured in populations of the cells using Fura-2AM and a charge coupled device (CCD) camera attached to a fluorescence microscope. dBcAMP induced both dose- and time-dependent changes in [ Ca(2+)](i) with increases in both the [Ca(2+)](i) oscillations and mean [Ca(2+)](i) (e.g. in C6 cells at 18 min mean [Ca(2+)](i) was 318 +/- 20nM following the single differentiating dBcAMP pulses, 489 +/- 17nM (p < 0.001) following two serial pulses, and 275 +/- 30nM (not significant) following three pulses). Therapeutic doses of fluoxetine and amitriptyline caused increases in the calcium oscillations and the mean calcium concentrations ( maximum recorded mean increase was in the C6 cells at 10min by 0.02 muM fluoxetine when [Ca(2+)](i) was 411 +/- 35nM c.f. control 254 +/- 25nM, p = 0.01). Higher (non-therapeutic) doses of both antidepressants caused significant reductions. Chloroquine and acrylamide also caused dose-dependent bi-phasic types of alterations in [Ca(2+)](i), with significant reductions at lower, sub-cytotoxic doses followed by significant increases at higher concentrations, approaching those which cause cell damage. Nifedipine treatment caused some reductions in the dBcAMP, antidepressant or toxicant-induced calcium changes, but this substance also initiated cytotoxic alterations. The findings show that both the activation-type changes (which are frequently associated with increased protective capacities) and toxic responses of C6 and 1321N1 cells to different chemical agents are associated with dose-dependent alterations in [Ca(2+)](i).
Collapse
Affiliation(s)
- N Debbie Slamon
- Division of Biosciences, University of Salford, Salford, U.K
| | | | | | | | | |
Collapse
|
41
|
Ostrow LW, Sachs F. Mechanosensation and endothelin in astrocytes--hypothetical roles in CNS pathophysiology. ACTA ACUST UNITED AC 2004; 48:488-508. [PMID: 15914254 DOI: 10.1016/j.brainresrev.2004.09.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2003] [Revised: 08/19/2004] [Accepted: 09/09/2004] [Indexed: 01/23/2023]
Abstract
Endothelin (ET) is a potent autocrine mitogen produced by reactive and neoplastic astrocytes. ET has been implicated in the induction of astrocyte proliferation and other transformations engendered by brain pathology, and in promoting the malignant behavior of astrocytomas. Reactive astrocytes containing ET are found in the periphery/penumbra of a wide array of CNS pathologies. Virtually all brain pathology deforms the surrounding parenchyma, either by direct mass effect or edema. Mechanical stress is a well established stimulus for ET production and release by other cell types, but has not been well studied in the brain. However, numerous studies have illustrated that astrocytes can sense mechanical stress and translate it into chemical messages. Furthermore, the ubiquitous reticular meshwork formed by interconnected astrocytes provides an ideal morphology for sensing and responding to mechanical disturbances. We have recently demonstrated stretch-induced ET production by astrocytes in vitro. Inspired by this finding, the purpose of this article is to review the literature on (1) astrocyte mechanosensation, and (2) the endothelin system in astrocytes, and to consider the hypothesis that mechanical induction of the ET system may influence astrocyte functioning in CNS pathophysiology. We conclude by discussing evidence supporting future investigations to determine whether specific inhibition of stretch-activated ion channels may represent a novel strategy for treating or preventing CNS disturbances, as well as the relevance to astrocyte-derived tumors.
Collapse
Affiliation(s)
- Lyle W Ostrow
- Department of Physiology and Biophysics, S.U.N.Y. at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA
| | | |
Collapse
|
42
|
Abstract
Abstract Although glia often envelop synapses, they have traditionally been viewed as passive participants in synaptic function. Recent evidence has demonstrated, however, that there is a dynamic two-way communication between glia and neurons at the synapse. Neurotransmitters released from presynaptic neurons evoke Ca2+ concentration increases in adjacent glia. Activated glia, in turn, release transmitters, including glutamate and ATP. These gliotransmitters feed back onto the presynaptic terminal either to enhance or to depress further release of neurotransmitter. Transmitters released from glia can also directly stimulate postsynaptic neurons, producing either excitatory or inhibitory responses. Based on these new findings, glia should be considered an active partner at the synapse, dynamically regulating synaptic transmission.
Collapse
Affiliation(s)
- Eric A Newman
- Department of Neuroscience, University of Minnesota, 6-145 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455, USA.
| |
Collapse
|
43
|
Abstract
ATP is a key extracellular messenger that mediates the propagation of Ca 2+ waves in astrocyte networks in various regions of the CNS. ATP-mediated Ca 2+ signals play critical roles in astrocyte proliferation and differentiation and in modulating neuronal activity. The actions of ATP on astrocytes are via two distinct subtypes of P2Y purinoceptors, P2Y1 and P2Y2 receptors (P2Y1Rs and P2Y2Rs), G-protein coupled receptors that stimulate mobilization of intracellular Ca 2+ ([Ca 2+]i) via the phospholipase Cbeta-IP3 pathway. We report here that P2Y1R-mediated and P2Y2R-mediated Ca 2+ responses differentially show two forms of activity-dependent negative feedback. First, Ca 2+ responses mediated by either receptor exhibit slow depression that is independent of stimulation frequency. Second, responses mediated by P2Y1Rs, but not those mediated by P2Y2Rs, show rapid oscillations after high-frequency stimulation. We demonstrate that the oscillations are mediated by recruiting negative feedback by protein kinase C, and we map the site responsible for the effect of protein kinase C to Thr339 in the C terminus of P2Y1R. We propose that frequency-dependent changes in ATP-mediated Ca 2+ signaling pathways may modulate astrocyte function and astrocyte-neuron signaling in the CNS.
Collapse
|
44
|
Berry CB, McBean GJ. An investigation into the role of calcium in the modulation of rat synaptosomal D-[3H]aspartate transport by docosahexaenoic acid. Brain Res 2003; 973:107-14. [PMID: 12729959 DOI: 10.1016/s0006-8993(03)02565-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The effect of the polyunsaturated fatty acid cis-4,7,10,13,16,19-docosahexaenoic acid (DHA) on the high-affinity, sodium-dependent uptake of D-[3H]aspartate into purified rat brain synaptosomes was examined. Incubation of the synaptosomes with 20 microM DHA caused over 50% inhibition of the maximum velocity (V(max)) of D-[3H]aspartate transport. This inhibition was significantly potentiated by pre-exposure of the synaptosomes to the fatty acid for 10 min prior to the start of the transport assay. Less highly unsaturated fatty acids such as arachidonic acid (cis-5,8,11,14-eicosatetraenoic acid), linolenic acid (cis-9,12,15-octadecatrienoic acid) and oleic acid (cis-9-octadecenoic acid) were significantly less potent than DHA. Removal of extracellular calcium, or reduction of the intracellular calcium concentration using the intracellular calcium chelator BAPTA/AM (10 microM), did not reduce the inhibition caused by DHA. On the other hand, an increase in the concentration of intracellular calcium mediated by thapsigargin (25 microM) or the calcium ionophore A23187 (10 or 100 nM) led to a reduction in the rate of D-[3H]aspartate transport in the absence of DHA. The CaM kinase II inhibitor, KN-93, reduced D-[3H]aspartate uptake independently of whether DHA was also present, but had no effect on the inhibition of D-[3H]aspartate uptake by either A23187 or thapsigargin. We conclude that whereas DHA inhibits synaptosomal D-[3H]aspartate uptake in a calcium-independent manner, a calcium-based mechanism exists that can also modulate glutamate transporter activity.
Collapse
Affiliation(s)
- Colm B Berry
- Department of Biochemistry, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | | |
Collapse
|
45
|
Sergeeva M, Strokin M, Wang H, Ubl JJ, Reiser G. Arachidonic acid in astrocytes blocks Ca(2+) oscillations by inhibiting store-operated Ca(2+) entry, and causes delayed Ca(2+) influx. Cell Calcium 2003; 33:283-92. [PMID: 12618149 DOI: 10.1016/s0143-4160(03)00011-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
ATP-elicited oscillations of the concentration of free intracellular Ca(2+) ([Ca(2+)](i)) in rat brain astrocytes were abolished by simultaneous arachidonic acid (AA) addition, whereas the tetraenoic analogue 5,8,11,14-eicosatetraynoic acid (ETYA) was ineffective. Inhibition of oscillations is due to suppression by AA of intracellular Ca(2+) store refilling. Short-term application of AA, but not ETYA, blocked Ca(2+) influx, which was evoked by depletion of stores with cyclopiazonic acid (CPA) or thapsigargin (Tg). Addition of AA after ATP blocked ongoing [Ca(2+)](i) oscillations. Prolonged AA application without or with agonist could evoke a delayed [Ca(2+)](i) increase. This AA-induced [Ca(2+)](i) rise developed slowly, reached a plateau after 5 min, could be reversed by addition of bovine serum albumin (BSA), that scavenges AA, and was blocked by 1 microM Gd(3+), indicative for the influx of extracellular Ca(2+). Specificity for AA as active agent was demonstrated by ineffectiveness of C16:0, C18:0, C20:0, C18:2, and ETYA. Moreover, the action of AA was not affected by inhibitors of oxidative metabolism of AA (ibuprofen, MK886, SKF525A). Thus, AA exerted a dual effect on astrocytic [Ca(2+)](i), firstly, a rapid reduction of capacitative Ca(2+) entry thereby suppressing [Ca(2+)](i) oscillations, and secondly inducing a delayed activation of Ca(2+) entry, also sensitive to low Gd(3+) concentration.
Collapse
Affiliation(s)
- Marina Sergeeva
- Institut für Neurobiochemie, Medizinische Fakultät der Otto-von-Guericke-Universität Magdeburg, Leipziger Strasse 44, D-39120 Magdeburg, Germany
| | | | | | | | | |
Collapse
|
46
|
Abstract
ATP is released by neurons and functions as a neurotransmitter and modulator in the CNS. Here I show that ATP released from glial cells can also serve as a potent neuromodulator, inhibiting neurons in the retina of the rat. Activation of glial cells by focal ejection of ATP, ATPgammaS, dopamine, thrombin, or lysophosphatidic acid or by mechanical stimulation evoked hyperpolarizing responses and outward currents in a subset of retinal ganglion cells by increasing a Ba(2+)-sensitive K(+) conductance in the neurons. This glia-evoked inhibition reduced the firing rate of those neurons that displayed spontaneous spike activity. The inhibition was abolished by the A(1) adenosine receptor antagonist DPCPX (8-cyclopentyl-1,3-dipropylxanthine) (10 nm) and was reduced by the ecto-ATPase inhibitor ARL-67156 (6-N,N-diethyl-D-beta,gamma-dibromomethyleneATP) (50 microm) and by the ectonucleotidase inhibitor AOPCP [adenosine-5'-O-(alpha,beta-methylene)-diphosphonate] (250 microm). Selective activation of retinal glial cells demonstrated that Müller cells, but not astrocytes, mediate the inhibition. ATP release from Müller cells into the inner plexiform layer of the retina was shown using the luciferin-luciferase chemiluminescence assay. These findings demonstrate that activated glial cells can inhibit neurons in the retina by the release of ATP, which is converted to adenosine by ectoenzymes and subsequently activates neuronal adenosine receptors. The results lend support to the hypothesis that glial cells play an active role in information processing in the CNS.
Collapse
|
47
|
Decoding calcium wave signaling. ACTA ACUST UNITED AC 2003. [DOI: 10.1016/s1569-2558(03)31030-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
48
|
Crunelli V, Blethyn KL, Cope DW, Hughes SW, Parri HR, Turner JP, Tòth TI, Williams SR. Novel neuronal and astrocytic mechanisms in thalamocortical loop dynamics. Philos Trans R Soc Lond B Biol Sci 2002; 357:1675-93. [PMID: 12626003 PMCID: PMC1693082 DOI: 10.1098/rstb.2002.1155] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In this review, we summarize three sets of findings that have recently been observed in thalamic astrocytes and neurons, and discuss their significance for thalamocortical loop dynamics. (i) A physiologically relevant 'window' component of the low-voltage-activated, T-type Ca(2+) current (I(Twindow)) plays an essential part in the slow (less than 1 Hz) sleep oscillation in adult thalamocortical (TC) neurons, indicating that the expression of this fundamental sleep rhythm in these neurons is not a simple reflection of cortical network activity. It is also likely that I(Twindow) underlies one of the cellular mechanisms enabling TC neurons to produce burst firing in response to novel sensory stimuli. (ii) Both electrophysiological and dye-injection experiments support the existence of gap junction-mediated coupling among young and adult TC neurons. This finding indicates that electrical coupling-mediated synchronization might be implicated in the high and low frequency oscillatory activities expressed by this type of thalamic neuron. (iii) Spontaneous intracellular Ca(2+) ([Ca(2+)](i)) waves propagating among thalamic astrocytes are able to elicit large and long-lasting N-methyl-D-aspartate-mediated currents in TC neurons. The peculiar developmental profile within the first two postnatal weeks of these astrocytic [Ca(2+)](i) transients and the selective activation of these glutamate receptors point to a role for this astrocyte-to-neuron signalling mechanism in the topographic wiring of the thalamocortical loop. As some of these novel cellular and intracellular properties are not restricted to thalamic astrocytes and neurons, their significance may well apply to (patho)physiological functions of glial and neuronal elements in other brain areas.
Collapse
Affiliation(s)
- Vincenzo Crunelli
- School of Biosciences, University of Cardiff, Museum Avenue, Cardiff CF10 3US, UK.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Two-way communication between neurons and nonneural cells called glia is essential for axonal conduction, synaptic transmission, and information processing and thus is required for normal functioning of the nervous system during development and throughout adult life. The signals between neurons and glia include ion fluxes, neurotransmitters, cell adhesion molecules, and specialized signaling molecules released from synaptic and nonsynaptic regions of the neuron. In contrast to the serial flow of information along chains of neurons, glia communicate with other glial cells through intracellular waves of calcium and via intercellular diffusion of chemical messengers. By releasing neurotransmitters and other extracellular signaling molecules, glia can affect neuronal excitability and synaptic transmission and perhaps coordinate activity across networks of neurons.
Collapse
Affiliation(s)
- R Douglas Fields
- Neurocytology and Physiology Section, National Institute of Child Health and Human Development, Bethesda, MD 20892, USA.
| | | |
Collapse
|
50
|
Yamane Y, Shiga H, Asou H, Ito E. GAP junctional channel inhibition alters actin organization and calcium propagation in rat cultured astrocytes. Neuroscience 2002; 112:593-603. [PMID: 12074901 DOI: 10.1016/s0306-4522(02)00095-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Astrocytes are connected by gap junctions, which provide intercellular pathways that allow a direct exchange of ions and small metabolites including second messengers and the propagation of electric currents. The roles of gap junctional communication on whole-cell morphology, cytoskeletal organization, and intercellular communication in astrocytes are not yet clear even in vitro, though there are many studies that have examined the active relation between gap junctions and actin filaments in astrocytes. Here we examined the effects of gap junction inhibitors, which do not interrupt the formation but rather the function of gap junctions, on whole-cell morphology, cytoskeletal organization, and intercellular communication in rat cultured astrocytes. Functional blockade of gap junctions during the formation of an astrocytic monolayer resulted in discordance of actin stress fibers between neighboring cells, even though whole-cell morphology of these cells did not change by such treatment. Mechanical stimulation-induced calcium wave propagation was significantly reduced in these actin-discordance cells even after thorough wash out. Differentiation of astrocytes in the presence of gap junction inhibitors was associated with morphological disarrangement among neighboring cells due to disordered alignment of actin stress fibers between cells.Our results indicate that gap junctional communication enables cell-to-cell coordination of actin stress fibers in astrocytes, thus enhancing intercellular communication through calcium spread.
Collapse
Affiliation(s)
- Y Yamane
- Division of Biological Sciences, Graduate School of Science, Hokkaido University, Sapporo, Japan
| | | | | | | |
Collapse
|