1
|
Muñoz-Ballester C, Robel S. Astrocyte-mediated mechanisms contribute to traumatic brain injury pathology. WIREs Mech Dis 2023; 15:e1622. [PMID: 37332001 PMCID: PMC10526985 DOI: 10.1002/wsbm.1622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 05/25/2023] [Accepted: 05/29/2023] [Indexed: 06/20/2023]
Abstract
Astrocytes respond to traumatic brain injury (TBI) with changes to their molecular make-up and cell biology, which results in changes in astrocyte function. These changes can be adaptive, initiating repair processes in the brain, or detrimental, causing secondary damage including neuronal death or abnormal neuronal activity. The response of astrocytes to TBI is often-but not always-accompanied by the upregulation of intermediate filaments, including glial fibrillary acidic protein (GFAP) and vimentin. Because GFAP is often upregulated in the context of nervous system disturbance, reactive astrogliosis is sometimes treated as an "all-or-none" process. However, the extent of astrocytes' cellular, molecular, and physiological adjustments is not equal for each TBI type or even for each astrocyte within the same injured brain. Additionally, new research highlights that different neurological injuries and diseases result in entirely distinctive and sometimes divergent astrocyte changes. Thus, extrapolating findings on astrocyte biology from one pathological context to another is problematic. We summarize the current knowledge about astrocyte responses specific to TBI and point out open questions that the field should tackle to better understand how astrocytes shape TBI outcomes. We address the astrocyte response to focal versus diffuse TBI and heterogeneity of reactive astrocytes within the same brain, the role of intermediate filament upregulation, functional changes to astrocyte function including potassium and glutamate homeostasis, blood-brain barrier maintenance and repair, metabolism, and reactive oxygen species detoxification, sex differences, and factors influencing astrocyte proliferation after TBI. This article is categorized under: Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Carmen Muñoz-Ballester
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Stefanie Robel
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
2
|
Benedikt J, Malpica-Nieves CJ, Rivera Y, Méndez-González M, Nichols CG, Veh RW, Eaton MJ, Skatchkov SN. The Polyamine Spermine Potentiates the Propagation of Negatively Charged Molecules through the Astrocytic Syncytium. Biomolecules 2022; 12:biom12121812. [PMID: 36551240 PMCID: PMC9775384 DOI: 10.3390/biom12121812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/16/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
The interest in astrocytes, the silent brain cells that accumulate polyamines (PAs), is growing. PAs exert anti-inflammatory, antioxidant, antidepressant, neuroprotective, and other beneficial effects, including increasing longevity in vivo. Unlike neurons, astrocytes are extensively coupled to others via connexin (Cx) gap junctions (GJs). Although there are striking modulatory effects of PAs on neuronal receptors and channels, PA regulation of the astrocytic GJs is not well understood. We studied GJ-propagation using molecules of different (i) electrical charge, (ii) structure, and (iii) molecular weight. Loading single astrocytes with patch pipettes containing membrane-impermeable dyes, we observed that (i) even small molecules do not easily permeate astrocytic GJs, (ii) the ratio of the charge to weight of these molecules is the key determinant of GJ permeation, (iii) the PA spermine (SPM) induced the propagation of negatively charged molecules via GJs, (iv) while no effects were observed on propagation of macromolecules with net-zero charge. The GJ uncoupler carbenoxolone (CBX) blocked such propagation. Taken together, these findings indicate that SPM is essential for astrocytic GJ communication and selectively facilitates intracellular propagation via GJs for negatively charged molecules through glial syncytium.
Collapse
Affiliation(s)
- Jan Benedikt
- Department of Physiology, Universidad Central del Caribe, Bayamón, PR 00956, USA
| | - Christian J. Malpica-Nieves
- Department of Biochemistry, Universidad Central del Caribe, Bayamón, PR 00956, USA
- Correspondence: (C.J.M.-N.); (S.N.S.); Tel.: +1-787-798-3001 (ext. 2057) (S.N.S.)
| | - Yomarie Rivera
- Department of Chiropractic, Universidad Central del Caribe, Bayamón, PR 00956, USA
| | | | - Colin G. Nichols
- Department of Cell Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rüdiger W. Veh
- Institut für Zell- und Neurobiologie, Charité, 10115 Berlin, Germany
| | - Misty J. Eaton
- Department of Biochemistry, Universidad Central del Caribe, Bayamón, PR 00956, USA
| | - Serguei N. Skatchkov
- Department of Physiology, Universidad Central del Caribe, Bayamón, PR 00956, USA
- Department of Biochemistry, Universidad Central del Caribe, Bayamón, PR 00956, USA
- Correspondence: (C.J.M.-N.); (S.N.S.); Tel.: +1-787-798-3001 (ext. 2057) (S.N.S.)
| |
Collapse
|
3
|
Velloso FJ, Shankar S, Parpura V, Rakic P, Levison SW. Neural Stem Cells in Adult Mammals are not Astrocytes. ASN Neuro 2022; 14:17590914221134739. [PMID: 36330653 PMCID: PMC9638700 DOI: 10.1177/17590914221134739] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/27/2022] [Accepted: 10/01/2022] [Indexed: 11/06/2022] Open
Abstract
At the turn of the 21st century studies of the cells that resided in the adult mammalian subventricular zone (SVZ) characterized the neural stem cells (NSCs) as a subtype of astrocyte. Over the ensuing years, numerous studies have further characterized the properties of these NSCs and compared them to parenchymal astrocytes. Here we have evaluated the evidence collected to date to establish whether classifying the NSCs as astrocytes is appropriate and useful. We also performed a meta-analysis with 4 previously published datasets that used cell sorting and unbiased single-cell RNAseq to highlight the distinct gene expression profiles of adult murine NSCs and niche astrocytes. On the basis of our understanding of the properties and functions of astrocytes versus the properties and functions of NSCs, and from our comparative transcriptomic analyses we conclude that classifying the adult mammalian NSC as an astrocyte is potentially misleading. From our vantage point, it is more appropriate to refer to the cells in the adult mammalian SVZ that retain the capacity to produce new neurons and macroglia as NSCs without attaching the term "astrocyte-like."
Collapse
Affiliation(s)
- Fernando Janczur Velloso
- Department of Pharmacology, Physiology & Neuroscience, New
Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Sandhya Shankar
- Department of Pharmacology, Physiology & Neuroscience, New
Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham,
Birmingham, AL, USA
| | - Pasko Rakic
- Department of Neuroscience, Yale School of Medicine, New Haven, CT,
USA
- Kavli Institute for Neuroscience, Yale School of Medicine, New
Haven, CT, USA
| | - Steven W. Levison
- Department of Pharmacology, Physiology & Neuroscience, New
Jersey Medical School, Rutgers University, Newark, NJ, USA
| |
Collapse
|
4
|
O'Donovan B, Neugornet A, Neogi R, Xia M, Ortinski P. Cocaine experience induces functional adaptations in astrocytes: Implications for synaptic plasticity in the nucleus accumbens shell. Addict Biol 2021; 26:e13042. [PMID: 33864336 DOI: 10.1111/adb.13042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 03/22/2021] [Accepted: 03/26/2021] [Indexed: 11/24/2022]
Abstract
Astrocytes have become established as an important regulator of neuronal activity in the brain. Accumulating literature demonstrates that cocaine self-administration in rodent models induces structural changes within astrocytes that may influence their interaction with the surrounding neurons. Here, we provide evidence that cocaine impacts astrocytes at the functional level and alters neuronal sensitivity to astrocyte-derived glutamate. We report that a 14-day period of short access to cocaine (2 h/day) decreases spontaneous astrocytic Ca2+ transients and precipitates changes in astrocyte network activity in the nucleus accumbens shell. This is accompanied by increased prevalence of slow inward currents, a physiological marker of neuronal activation by astrocytic glutamate, in a subset of medium spiny neurons. Within, but not outside, of this subset, we observe an increase in duration and frequency of N-methyl-D-aspartate (NMDA) receptor-mediated synaptic events. Additionally, we find that the link between synaptic NMDA receptor plasticity and neuron sensitivity to astrocytic glutamate is maintained independent of drug exposure and is observed in both cocaine and saline control animals. Imaging analyses of neuronal Ca2+ activity show no effect of cocaine self-administration on individual cells or on neuronal network activity in brain slices. Therefore, our data indicate that cocaine self-administration promotes astrocyte-specific functional changes that can be linked to increased glutamate-mediated coupling with principal neurons in the nucleus accumbens. Such coupling may be spatially restricted as it does not result in a broad impact on network structure of local neuronal circuits.
Collapse
Affiliation(s)
- Bernadette O'Donovan
- Department of Neuroscience, College of Medicine University of Kentucky Lexington Kentucky USA
| | - Austin Neugornet
- Department of Neuroscience, College of Medicine University of Kentucky Lexington Kentucky USA
| | - Richik Neogi
- Department of Neuroscience, College of Medicine University of Kentucky Lexington Kentucky USA
- Integrated Biomedical Sciences University of Kentucky Lexington Kentucky USA
| | - Mengfan Xia
- Department of Neuroscience, College of Medicine University of Kentucky Lexington Kentucky USA
| | - Pavel Ortinski
- Department of Neuroscience, College of Medicine University of Kentucky Lexington Kentucky USA
| |
Collapse
|
5
|
In Vivo Neocortical [K]o Modulation by Targeted Stimulation of Astrocytes. Int J Mol Sci 2021; 22:ijms22168658. [PMID: 34445364 PMCID: PMC8395460 DOI: 10.3390/ijms22168658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/05/2021] [Accepted: 08/09/2021] [Indexed: 11/17/2022] Open
Abstract
A normally functioning nervous system requires normal extracellular potassium ion concentration ([K]o). Throughout the nervous system, several processes, including those of an astrocytic nature, are involved in [K]o regulation. In this study we investigated the effect of astrocytic photostimulation on [K]o. We hypothesized that in vivo photostimulation of eNpHR-expressing astrocytes leads to a decreased [K]o. Using optogenetic and electrophysiological techniques we showed that stimulation of eNpHR-expressing astrocytes resulted in a significantly decreased resting [K]o and evoked K responses. The amplitude of the concomitant spreading depolarization-like events also decreased. Our results imply that astrocytic membrane potential modification could be a potential tool for adjusting the [K]o.
Collapse
|
6
|
Perez C, Felix L, Durry S, Rose CR, Ullah G. On the origin of ultraslow spontaneous Na + fluctuations in neurons of the neonatal forebrain. J Neurophysiol 2021; 125:408-425. [PMID: 33236936 PMCID: PMC7948148 DOI: 10.1152/jn.00373.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 11/19/2020] [Accepted: 11/19/2020] [Indexed: 11/22/2022] Open
Abstract
Spontaneous neuronal and astrocytic activity in the neonate forebrain is believed to drive the maturation of individual cells and their integration into complex brain-region-specific networks. The previously reported forms include bursts of electrical activity and oscillations in intracellular Ca2+ concentration. Here, we use ratiometric Na+ imaging to demonstrate spontaneous fluctuations in the intracellular Na+ concentration of CA1 pyramidal neurons and astrocytes in tissue slices obtained from the hippocampus of mice at postnatal days 2-4 (P2-4). These occur at very low frequency (∼2/h), can last minutes with amplitudes up to several millimolar, and mostly disappear after the first postnatal week. To further investigate their mechanisms, we model a network consisting of pyramidal neurons and interneurons. Experimentally observed Na+ fluctuations are mimicked when GABAergic inhibition in the simulated network is made depolarizing. Both our experiments and computational model show that blocking voltage-gated Na+ channels or GABAergic signaling significantly diminish the neuronal Na+ fluctuations. On the other hand, blocking a variety of other ion channels, receptors, or transporters including glutamatergic pathways does not have significant effects. Our model also shows that the amplitude and duration of Na+ fluctuations decrease as we increase the strength of glial K+ uptake. Furthermore, neurons with smaller somatic volumes exhibit fluctuations with higher frequency and amplitude. As opposed to this, larger extracellular to intracellular volume ratio observed in neonatal brain exerts a dampening effect. Finally, our model predicts that these periods of spontaneous Na+ influx leave neonatal neuronal networks more vulnerable to seizure-like states when compared with mature brain.NEW & NOTEWORTHY Spontaneous activity in the neonate forebrain plays a key role in cell maturation and brain development. We report spontaneous, ultraslow, asynchronous fluctuations in the intracellular Na+ concentration of neurons and astrocytes. We show that this activity is not correlated with the previously reported synchronous neuronal population bursting or Ca2+ oscillations, both of which occur at much faster timescales. Furthermore, extracellular K+ concentration remains nearly constant. The spontaneous Na+ fluctuations disappear after the first postnatal week.
Collapse
Affiliation(s)
- Carlos Perez
- Department of Physics, University of South Florida, Tampa, Florida
| | - Lisa Felix
- Faculty of Mathematics and Natural Sciences, Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Simone Durry
- Faculty of Mathematics and Natural Sciences, Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christine R Rose
- Faculty of Mathematics and Natural Sciences, Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ghanim Ullah
- Department of Physics, University of South Florida, Tampa, Florida
| |
Collapse
|
7
|
Götz S, Bribian A, López-Mascaraque L, Götz M, Grothe B, Kunz L. Heterogeneity of astrocytes: Electrophysiological properties of juxtavascular astrocytes before and after brain injury. Glia 2020; 69:346-361. [PMID: 32809228 DOI: 10.1002/glia.23900] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 07/20/2020] [Accepted: 07/30/2020] [Indexed: 12/31/2022]
Abstract
Astrocyte heterogeneity is increasingly recognized, but still little is known about juxtavascular astrocytes with their somata directly adjacent to blood vessels, despite their importance after brain injury. As juxtavascular astrocytes originate from common progenitor cells, that is, have a clonal origin, they may intrinsically differ from other, non-juxtavascular astrocytes. To explore this, we examined the electrophysiological properties of these groups of astrocytes and the underlying ion channels. Using brain slices of BAC Aldh1l1-eGFP transgenic mice with astrocytes labeled by GFP expression, we compared juxtavascular and non-juxtavascular astrocytes in the somatosensory cortex by means of whole-cell patch-clamp recordings and immunohistochemical staining. Prior to injury, juxta- and non-juxtavascular astrocytes exhibit comparable electrophysiological properties with characteristic mostly passive conductance and a typical negative resting membrane potential. Immunohistochemical analysis of K+ channels showed that all astrocytes were Kir 4.1+ , but revealed an intriguing difference for Kv 4.3. The expression of Kv 4.3 in sibling astrocytes (non-juxtavascular, juxtavascular and pial) was dependent on their ontogenetic origin with lowest levels in juxtavascular astrocytes located in upper cortical layers. After traumatic brain injury (TBI), we found profound changes in the electrophysiological type of astrocytes with a predominance of non-passive properties and this pattern was significantly enriched in juxtavascular astrocytes. This was accompanied by pronounced down-regulation of Kir 4.1 in proliferating astrocytes, which was significantly more in juxtavascular compared to non-juxtavascular astrocytes. Taken together, TBI induces profound differences in electrophysiological properties between juxtavascular and non-juxtavascular astrocytes that might be related to the preponderance of juxtavascular astrocyte proliferation.
Collapse
Affiliation(s)
- Stefanie Götz
- Division of Neurobiology, Department of Biology II, Ludwig-Maximilians-Universitaet (LMU) Munich, Martinsried, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universitaet (LMU) Munich, Martinsried, Germany.,Biomedical Center (BMC), Ludwig-Maximilians-Universitaet (LMU) Munich, SyNergy - Munich Cluster for Systems Neurology, Munich, Germany
| | - Ana Bribian
- Instituto Cajal-CSIC, Molecular, Cellular and Developmental Neurobiology Department, Madrid, Spain
| | - Laura López-Mascaraque
- Instituto Cajal-CSIC, Molecular, Cellular and Developmental Neurobiology Department, Madrid, Spain
| | - Magdalena Götz
- Biomedical Center (BMC), Ludwig-Maximilians-Universitaet (LMU) Munich, SyNergy - Munich Cluster for Systems Neurology, Munich, Germany.,Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians-Universitaet (LMU) Munich, Martinsried, Germany.,Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, Martinsried, Germany
| | - Benedikt Grothe
- Division of Neurobiology, Department of Biology II, Ludwig-Maximilians-Universitaet (LMU) Munich, Martinsried, Germany.,Biomedical Center (BMC), Ludwig-Maximilians-Universitaet (LMU) Munich, SyNergy - Munich Cluster for Systems Neurology, Munich, Germany
| | - Lars Kunz
- Division of Neurobiology, Department of Biology II, Ludwig-Maximilians-Universitaet (LMU) Munich, Martinsried, Germany
| |
Collapse
|
8
|
Felix L, Stephan J, Rose CR. Astrocytes of the early postnatal brain. Eur J Neurosci 2020; 54:5649-5672. [PMID: 32406559 DOI: 10.1111/ejn.14780] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/30/2020] [Accepted: 05/06/2020] [Indexed: 12/21/2022]
Abstract
In the rodent forebrain, the majority of astrocytes are generated during the early postnatal phase. Following differentiation, astrocytes undergo maturation which accompanies the development of the neuronal network. Neonate astrocytes exhibit a distinct morphology and domain size which differs to their mature counterparts. Moreover, many of the plasma membrane proteins prototypical for fully developed astrocytes are only expressed at low levels at neonatal stages. These include connexins and Kir4.1, which define the low membrane resistance and highly negative membrane potential of mature astrocytes. Newborn astrocytes moreover express only low amounts of GLT-1, a glutamate transporter critical later in development. Furthermore, they show specific differences in the properties and spatio-temporal pattern of intracellular calcium signals, resulting from differences in their repertoire of receptors and signalling pathways. Therefore, roles fulfilled by mature astrocytes, including ion and transmitter homeostasis, are underdeveloped in the young brain. Similarly, astrocytic ion signalling in response to neuronal activity, a process central to neuron-glia interaction, differs between the neonate and mature brain. This review describes the unique functional properties of astrocytes in the first weeks after birth and compares them to later stages of development. We conclude that with an immature neuronal network and wider extracellular space, astrocytic support might not be as demanding and critical compared to the mature brain. The delayed differentiation and maturation of astrocytes in the first postnatal weeks might thus reflect a reduced need for active, energy-consuming regulation of the extracellular space and a less tight control of glial feedback onto synaptic transmission.
Collapse
Affiliation(s)
- Lisa Felix
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Jonathan Stephan
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Christine R Rose
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
9
|
Spontaneous Ultraslow Na + Fluctuations in the Neonatal Mouse Brain. Cells 2019; 9:cells9010102. [PMID: 31906100 PMCID: PMC7016939 DOI: 10.3390/cells9010102] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/18/2019] [Accepted: 12/24/2019] [Indexed: 12/12/2022] Open
Abstract
In the neonate forebrain, network formation is driven by the spontaneous synchronized activity of pyramidal cells and interneurons, consisting of bursts of electrical activity and intracellular Ca2+ oscillations. By employing ratiometric Na+ imaging in tissue slices obtained from animals at postnatal day 2-4 (P2-4), we found that 20% of pyramidal neurons and 44% of astrocytes in neonatal mouse hippocampus also exhibit transient fluctuations in intracellular Na+. These occurred at very low frequencies (~2/h), were exceptionally long (~8 min), and strongly declined after the first postnatal week. Similar Na+ fluctuations were also observed in the neonate neocortex. In the hippocampus, Na+ elevations in both cell types were diminished when blocking action potential generation with tetrodotoxin. Neuronal Na+ fluctuations were significantly reduced by bicuculline, suggesting the involvement of GABAA-receptors in their generation. Astrocytic signals, by contrast, were neither blocked by inhibition of receptors and/or transporters for different transmitters including GABA and glutamate, nor of various Na+-dependent transporters or Na+-permeable channels. In summary, our results demonstrate for the first time that neonatal astrocytes and neurons display spontaneous ultraslow Na+ fluctuations. While neuronal Na+ signals apparently largely rely on suprathreshold GABAergic excitation, astrocytic Na+ signals, albeit being dependent on neuronal action potentials, appear to have a separate trigger and mechanism, the source of which remains unclear at present.
Collapse
|
10
|
Bedner P, Jabs R, Steinhäuser C. Properties of human astrocytes and NG2 glia. Glia 2019; 68:756-767. [DOI: 10.1002/glia.23725] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/09/2019] [Accepted: 08/09/2019] [Indexed: 01/11/2023]
Affiliation(s)
- Peter Bedner
- Institute of Cellular Neurosciences, Medical FacultyUniversity of Bonn Bonn Germany
| | - Ronald Jabs
- Institute of Cellular Neurosciences, Medical FacultyUniversity of Bonn Bonn Germany
| | | |
Collapse
|
11
|
Postnatal Increases in Axonal Conduction Velocity of an Identified Drosophila Interneuron Require Fast Sodium, L-Type Calcium and Shaker Potassium Channels. eNeuro 2019; 6:ENEURO.0181-19.2019. [PMID: 31253715 PMCID: PMC6709211 DOI: 10.1523/eneuro.0181-19.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/06/2019] [Accepted: 06/12/2019] [Indexed: 11/21/2022] Open
Abstract
During early postnatal life, speed up of signal propagation through many central and peripheral neurons has been associated with an increase in axon diameter or/and myelination. Especially in unmyelinated axons postnatal adjustments of axonal membrane conductances is potentially a third mechanism but solid evidence is lacking. Here, we show that axonal action potential (AP) conduction velocity in the Drosophila giant fiber (GF) interneuron, which is required for fast long-distance signal conduction through the escape circuit, is increased by 80% during the first day of adult life. Genetic manipulations indicate that this postnatal increase in AP conduction velocity in the unmyelinated GF axon is likely owed to adjustments of ion channel expression or properties rather than axon diameter increases. Specifically, targeted RNAi knock-down of either Para fast voltage-gated sodium, Shaker potassium (Kv1 homologue), or surprisingly, L-type like calcium channels counteracts postnatal increases in GF axonal conduction velocity. By contrast, the calcium-dependent potassium channel Slowpoke (BK) is not essential for postnatal speeding, although it also significantly increases conduction velocity. Therefore, we identified multiple ion channels that function to support fast axonal AP conduction velocity, but only a subset of these are regulated during early postnatal life to maximize conduction velocity. Despite its large diameter (∼7 µm) and postnatal regulation of multiple ionic conductances, mature GF axonal conduction velocity is still 20-60 times slower than that of vertebrate Aβ sensory axons and α motoneurons, thus unraveling the limits of long-range information transfer speed through invertebrate circuits.
Collapse
|
12
|
Garcia VJ, Rushton DJ, Tom CM, Allen ND, Kemp PJ, Svendsen CN, Mattis VB. Huntington's Disease Patient-Derived Astrocytes Display Electrophysiological Impairments and Reduced Neuronal Support. Front Neurosci 2019; 13:669. [PMID: 31316341 PMCID: PMC6610155 DOI: 10.3389/fnins.2019.00669] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/11/2019] [Indexed: 12/03/2022] Open
Abstract
In Huntington’s disease (HD), while the ubiquitously expressed mutant Huntingtin (mtHTT) protein primarily compromises striatal and cortical neurons, glia also undergo disease-contributing alterations. Existing HD models using human induced pluripotent stem cells (iPSCs) have not extensively characterized the role of mtHTT in patient-derived astrocytes. Here physiologically mature astrocytes are generated from HD patient iPSCs. These human astrocytes exhibit hallmark HD phenotypes that occur in mouse models, including impaired inward rectifying K+ currents, lengthened spontaneous Ca2+ waves and reduced cell membrane capacitance. HD astrocytes in co-culture provided reduced support for the maturation of iPSC-derived neurons. In addition, neurons exposed to chronic glutamate stimulation are not protected by HD astrocytes. This iPSC-based HD model demonstrates the critical effects of mtHTT on human astrocytes, which not only broadens the understanding of disease susceptibility beyond cortical and striatal neurons but also increases potential drug targets.
Collapse
Affiliation(s)
- Veronica J Garcia
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - David J Rushton
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Divisions of Biomedicine and Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Colton M Tom
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Nicholas D Allen
- Divisions of Biomedicine and Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Paul J Kemp
- Divisions of Biomedicine and Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Virginia B Mattis
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
13
|
Li AQ, Sun ZP, Liu X, Yang JS, Jin F, Zhu L, Jia WH, De Vos S, Van Stappen G, Bossier P, Yang WJ. The chloride channel cystic fibrosis transmembrane conductance regulator (CFTR) controls cellular quiescence by hyperpolarizing the cell membrane during diapause in the crustacean Artemia. J Biol Chem 2019; 294:6598-6611. [PMID: 30765604 DOI: 10.1074/jbc.ra118.005900] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 02/10/2019] [Indexed: 01/10/2023] Open
Abstract
Cellular quiescence, a reversible state in which growth, proliferation, and other cellular activities are arrested, is important for self-renewal, differentiation, development, regeneration, and stress resistance. However, the physiological mechanisms underlying cellular quiescence remain largely unknown. In the present study, we used embryos of the crustacean Artemia in the diapause stage, in which these embryos remain quiescent for prolonged periods, as a model to explore the relationship between cell-membrane potential (V mem) and quiescence. We found that V mem is hyperpolarized and that the intracellular chloride concentration is high in diapause embryos, whereas V mem is depolarized and intracellular chloride concentration is reduced in postdiapause embryos and during further embryonic development. We identified and characterized the chloride ion channel protein cystic fibrosis transmembrane conductance regulator (CFTR) of Artemia (Ar-CFTR) and found that its expression is silenced in quiescent cells of Artemia diapause embryos but remains constant in all other embryonic stages. Ar-CFTR knockdown and GlyH-101-mediated chemical inhibition of Ar-CFTR produced diapause embryos having a high V mem and intracellular chloride concentration, whereas control Artemia embryos released free-swimming nauplius larvae. Transcriptome analysis of embryos at different developmental stages revealed that proliferation, differentiation, and metabolism are suppressed in diapause embryos and restored in postdiapause embryos. Combined with RNA sequencing (RNA-Seq) of GlyH-101-treated MCF-7 breast cancer cells, these analyses revealed that CFTR inhibition down-regulates the Wnt and Aurora Kinase A (AURKA) signaling pathways and up-regulates the p53 signaling pathway. Our findings provide insight into CFTR-mediated regulation of cellular quiescence and V mem in the Artemia model.
Collapse
Affiliation(s)
- An-Qi Li
- From the College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Zhan-Peng Sun
- From the College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xu Liu
- From the College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jin-Shu Yang
- From the College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Feng Jin
- From the College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Lin Zhu
- From the College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Wen-Huan Jia
- From the College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Stephanie De Vos
- the Laboratory of Aquaculture and Artemia Reference Center, Department of Animal Production, Ghent University, B-9000 Ghent, Belgium, and
| | - Gilbert Van Stappen
- the Laboratory of Aquaculture and Artemia Reference Center, Department of Animal Production, Ghent University, B-9000 Ghent, Belgium, and
| | - Peter Bossier
- the Laboratory of Aquaculture and Artemia Reference Center, Department of Animal Production, Ghent University, B-9000 Ghent, Belgium, and
| | - Wei-Jun Yang
- From the College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China, .,the Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266000, China
| |
Collapse
|
14
|
Heterogeneity of Activity-Induced Sodium Transients between Astrocytes of the Mouse Hippocampus and Neocortex: Mechanisms and Consequences. J Neurosci 2019; 39:2620-2634. [PMID: 30737311 DOI: 10.1523/jneurosci.2029-18.2019] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 01/07/2019] [Accepted: 01/23/2019] [Indexed: 01/09/2023] Open
Abstract
Activity-related sodium transients induced by glutamate uptake represent a special form of astrocyte excitability. Astrocytes of the neocortex, as opposed to the hippocampus proper, also express ionotropic glutamate receptors, which might provide additional sodium influx. We compared glutamate-related sodium transients in astrocytes and neurons in slices of the neocortex and hippocampus of juvenile mice of both sexes, using widefield and multiphoton imaging. Stimulation of glutamatergic afferents or glutamate application induced sodium transients that were twice as large in neocortical as in hippocampal astrocytes, despite similar neuronal responses. Astrocyte sodium transients were reduced by ∼50% upon blocking NMDA receptors in the neocortex, but not hippocampus. Neocortical, but not hippocampal, astrocytes exhibited marked sodium increases in response to NMDA. These key differences in sodium signaling were also observed in neonates and in adults. NMDA application evoked local calcium transients in processes of neocortical astrocytes, which were dampened upon blocking sodium/calcium exchange (NCX) with KB-R7943 or SEA0400. Mathematical computation based on our data predict that NMDA-induced sodium increases drive the NCX into reverse mode, resulting in calcium influx. Together, our study reveals a considerable regional heterogeneity in astrocyte sodium transients, which persists throughout postnatal development. Neocortical astrocytes respond with much larger sodium elevations to glutamatergic activity than hippocampal astrocytes. Moreover, neocortical astrocytes experience NMDA-receptor-mediated sodium influx, which hippocampal astrocytes lack, and which drives calcium import through reverse NCX. This pathway thereby links sodium to calcium signaling and represents a new mechanism for the generation of local calcium influx in neocortical astrocytes.SIGNIFICANCE STATEMENT Astrocyte calcium signals play a central role in neuron-glia interaction. Moreover, activity-related sodium transients may represent a new form of astrocyte excitability. Here we show that activation of NMDA receptors results in prominent sodium transients in neocortical, but not hippocampal, astrocytes in the mouse brain. NMDA receptor activation is accompanied by local calcium signaling in processes of neocortical astrocytes, which is augmented by sodium-driven reversal of the sodium/calcium exchanger. Our data demonstrate a significant regional heterogeneity in the magnitude and mechanisms of astrocyte sodium transients. They also suggest a close interrelation between NMDA-receptor-mediated sodium influx and calcium signaling through the reversal of sodium/calcium exchanger, thereby establishing a new pathway for the generation of local calcium signaling in astrocyte processes.
Collapse
|
15
|
Larson VA, Mironova Y, Vanderpool KG, Waisman A, Rash JE, Agarwal A, Bergles DE. Oligodendrocytes control potassium accumulation in white matter and seizure susceptibility. eLife 2018; 7:34829. [PMID: 29596047 PMCID: PMC5903864 DOI: 10.7554/elife.34829] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/28/2018] [Indexed: 12/19/2022] Open
Abstract
The inwardly rectifying K+ channel Kir4.1 is broadly expressed by CNS glia and deficits in Kir4.1 lead to seizures and myelin vacuolization. However, the role of oligodendrocyte Kir4.1 channels in controlling myelination and K+ clearance in white matter has not been defined. Here, we show that selective deletion of Kir4.1 from oligodendrocyte progenitors (OPCs) or mature oligodendrocytes did not impair their development or disrupt the structure of myelin. However, mice lacking oligodendrocyte Kir4.1 channels exhibited profound functional impairments, including slower clearance of extracellular K+ and delayed recovery of axons from repetitive stimulation in white matter, as well as spontaneous seizures, a lower seizure threshold, and activity-dependent motor deficits. These results indicate that Kir4.1 channels in oligodendrocytes play an important role in extracellular K+ homeostasis in white matter, and that selective loss of this channel from oligodendrocytes is sufficient to impair K+ clearance and promote seizures.
Collapse
Affiliation(s)
- Valerie A Larson
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Yevgeniya Mironova
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Kimberly G Vanderpool
- Department of Biomedical Sciences, Colorado State University, Fort Collins, United States
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - John E Rash
- Department of Biomedical Sciences, Colorado State University, Fort Collins, United States
| | - Amit Agarwal
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Dwight E Bergles
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| |
Collapse
|
16
|
Heterogeneity and function of hippocampal macroglia. Cell Tissue Res 2017; 373:653-670. [PMID: 29204745 DOI: 10.1007/s00441-017-2746-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 11/16/2017] [Indexed: 12/26/2022]
Abstract
The contribution of glial cells to normal and impaired hippocampal function is increasingly being recognized, although important questions as to the mechanisms that these cells use for their crosstalk with neurons and capillaries are still unanswered or lead to controversy. Astrocytes in the hippocampus are morphologically variable and a single cell contacts with its processes more than 100,000 synapses. They predominantly express inward rectifier K+ channels and transporters serving homeostatic function but may also release gliotransmitters to modify neuronal signaling and brain circulation. Intracellular Ca2+ transients are key events in the interaction of astrocytes with neurons and the vasculature. Hippocampal NG2 glia represent a population of cells with proliferative capacity throughout adulthood. Intriguingly, they receive direct synaptic input from pyramidal neurons and interneurons and express a multitude of ion channels and receptors. Despite in-depth knowledge about the features of these transmembrane proteins, the physiological impact of NG2 glial cells and their synaptic input remain nebulous. Because of the low abundance of oligodendrocytes in the hippocampus, limited information is available about their specific properties. Given the multitude of signaling molecules expressed by the various types of hippocampal glial cells (and because of space constraints), we focus, in this review, on those properties that are considered key for the interaction of the respective cell type with its neighborhood.
Collapse
|
17
|
Rose CR, Ziemens D, Untiet V, Fahlke C. Molecular and cellular physiology of sodium-dependent glutamate transporters. Brain Res Bull 2016; 136:3-16. [PMID: 28040508 DOI: 10.1016/j.brainresbull.2016.12.013] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/20/2016] [Accepted: 12/21/2016] [Indexed: 02/04/2023]
Abstract
Glutamate is the major excitatory transmitter in the vertebrate brain. After its release from presynaptic nerve terminals, it is rapidly taken up by high-affinity sodium-dependent plasma membrane transporters. While both neurons and glial cells express these excitatory amino acid transporters (EAATs), the majority of glutamate uptake is accomplished by astrocytes, which convert synaptically-released glutamate to glutamine or feed it into their own metabolism. Glutamate uptake by astrocytes not only shapes synaptic transmission by regulating the availability of glutamate to postsynaptic neuronal receptors, but also protects neurons from hyper-excitability and subsequent excitotoxic damage. In the present review, we provide an overview of the molecular and cellular characteristics of sodium-dependent glutamate transporters and their associated anion permeation pathways, with a focus on astrocytic glutamate transport. We summarize their functional properties and roles within tripartite synapses under physiological and pathophysiological conditions, exemplifying the intricate interactions and interrelationships between neurons and glial cells in the brain.
Collapse
Affiliation(s)
- Christine R Rose
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Germany.
| | - Daniel Ziemens
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Germany
| | - Verena Untiet
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, Germany
| | - Christoph Fahlke
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, Germany
| |
Collapse
|
18
|
Untiet V, Kovermann P, Gerkau NJ, Gensch T, Rose CR, Fahlke C. Glutamate transporter-associated anion channels adjust intracellular chloride concentrations during glial maturation. Glia 2016; 65:388-400. [PMID: 27859594 DOI: 10.1002/glia.23098] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 10/18/2016] [Accepted: 10/24/2016] [Indexed: 01/09/2023]
Abstract
Astrocytic volume regulation and neurotransmitter uptake are critically dependent on the intracellular anion concentration, but little is known about the mechanisms controlling internal anion homeostasis in these cells. Here we used fluorescence lifetime imaging microscopy (FLIM) with the chloride-sensitive dye MQAE to measure intracellular chloride concentrations in murine Bergmann glial cells in acute cerebellar slices. We found Bergmann glial [Cl- ]int to be controlled by two opposing transport processes: chloride is actively accumulated by the Na+ -K+ -2Cl- cotransporter NKCC1, and chloride efflux through anion channels associated with excitatory amino acid transporters (EAATs) reduces [Cl- ]int to values that vary upon changes in expression levels or activity of these channels. EAATs transiently form anion-selective channels during glutamate transport, and thus represent a class of ligand-gated anion channels. Age-dependent upregulation of EAATs results in a developmental chloride switch from high internal chloride concentrations (51.6 ± 2.2 mM, mean ± 95% confidence interval) during early development to adult levels (35.3 ± 0.3 mM). Simultaneous blockade of EAAT1/GLAST and EAAT2/GLT-1 increased [Cl- ]int in adult glia to neonatal values. Moreover, EAAT activation by synaptic stimulations rapidly decreased [Cl- ]int . Other tested chloride channels or chloride transporters do not contribute to [Cl- ]int under our experimental conditions. Neither genetic removal of ClC-2 nor pharmacological block of K+ -Cl- cotransporter change resting Bergmann glial [Cl- ]int in acute cerebellar slices. We conclude that EAAT anion channels play an important and unexpected role in adjusting glial intracellular anion concentration during maturation and in response to cerebellar activity. GLIA 2017;65:388-400.
Collapse
Affiliation(s)
- Verena Untiet
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, Germany
| | - Peter Kovermann
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, Germany
| | - Niklas J Gerkau
- Institute of Neurobiology, Heinrich-Heine-Universität Düsseldorf, Germany
| | - Thomas Gensch
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, Germany
| | - Christine R Rose
- Institute of Neurobiology, Heinrich-Heine-Universität Düsseldorf, Germany
| | - Christoph Fahlke
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, Germany
| |
Collapse
|
19
|
Zhou N, Huang S, Li L, Huang D, Yan Y, Du X, Zhang H. Suppression of KV7/KCNQ potassium channel enhances neuronal differentiation of PC12 cells. Neuroscience 2016; 333:356-67. [PMID: 27450567 DOI: 10.1016/j.neuroscience.2016.07.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 07/07/2016] [Accepted: 07/08/2016] [Indexed: 11/28/2022]
Abstract
Membrane potential shift driven by electrical activity is critical in determining the cell fate of proliferation or differentiation. As such, the ion channels that underlie the membrane electrical activity play an important role in cell proliferation/differentiation. KV7/KCNQ potassium channels are critical in determining the resting membrane potentials in many neuronal cells. However, the role of these channels in cell differentiation is not well studied. In the present study, we used PC12 cells as well as primary cultured rat cortical neurons to study the role and mechanism of KV7/KCNQ in neuronal differentiation. NGF induced PC12 cell differentiation into neuron-like cells with growth of neurites showing typical growth cone-like extensions. The Kv7/KCNQ blocker XE991 promoted NGF-induced neurite outgrowth, whereas Kv7/KCNQ opener retigabine (RTG) inhibited outgrowth. M-type Kv7 channels are likely involved in regulating neurite growth because overexpression of KCNQ2/Q3 inhibited neurite growth whereas suppression of KCNQ2/Q3 with shRNA promoted neurite growth. Membrane depolarization possibly underpins enhanced neurite growth induced by the suppression of Kv7/KCNQ. Additionally, high extracellular K(+) likely induced membrane depolarization and also promoted neurite growth. Finally, T-type Ca(2+) channels may be involved in membrane-depolarization-induced neurite growth. This study provides a new perspective for understanding neuronal differentiation as well as KV7/KCNQ channel function.
Collapse
Affiliation(s)
- Najing Zhou
- Department of Pharmacology, Hebei Medical University, China; Department of Cell Biology, Hebei Medical University, Shijiazhuang 050017, China
| | - Sha Huang
- Department of Pharmacology, Hebei Medical University, China
| | - Li Li
- Department of Pharmacology, Hebei Medical University, China
| | - Dongyang Huang
- Department of Pharmacology, Hebei Medical University, China
| | - Yunli Yan
- Department of Cell Biology, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiaona Du
- Department of Pharmacology, Hebei Medical University, China
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical University, China.
| |
Collapse
|
20
|
Abstract
Epilepsy is among the most prevalent chronic neurological diseases and affects an estimated 2.2 million people in the United States alone. About one third of patients are resistant to currently available antiepileptic drugs, which are exclusively targeting neuronal function. Yet, reactive astrocytes have emerged as potential contributors to neuronal hyperexcitability and seizures. Astrocytes react to any kind of CNS insult with a range of cellular adjustments to form a scar and protect uninjured brain regions. This process changes astrocyte physiology and can affect neuronal network function in various ways. Traumatic brain injury and stroke, both conditions that trigger astroglial scar formation, are leading causes of acquired epilepsies and surgical removal of this glial scar in patients with drug-resistant epilepsy can alleviate the seizures. This review will summarize the currently available evidence suggesting that epilepsy is not a disease of neurons alone, but that astrocytes, glial cells in the brain, can be major contributors to the disease, especially when they adopt a reactive state in response to central nervous system insult.
Collapse
Affiliation(s)
- Stefanie Robel
- Virginia Tech Carilion Research Institute, Roanoke, VA, USA
- Virginia Tech School of Neuroscience, Blacksburg, VA, USA
| |
Collapse
|
21
|
Nwaobi SE, Cuddapah VA, Patterson KC, Randolph AC, Olsen ML. The role of glial-specific Kir4.1 in normal and pathological states of the CNS. Acta Neuropathol 2016; 132:1-21. [PMID: 26961251 PMCID: PMC6774634 DOI: 10.1007/s00401-016-1553-1] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 02/16/2016] [Accepted: 02/25/2016] [Indexed: 12/15/2022]
Abstract
Kir4.1 is an inwardly rectifying K(+) channel expressed exclusively in glial cells in the central nervous system. In glia, Kir4.1 is implicated in several functions including extracellular K(+) homeostasis, maintenance of astrocyte resting membrane potential, cell volume regulation, and facilitation of glutamate uptake. Knockout of Kir4.1 in rodent models leads to severe neurological deficits, including ataxia, seizures, sensorineural deafness, and early postnatal death. Accumulating evidence indicates that Kir4.1 plays an integral role in the central nervous system, prompting many laboratories to study the potential role that Kir4.1 plays in human disease. In this article, we review the growing evidence implicating Kir4.1 in a wide array of neurological disease. Recent literature suggests Kir4.1 dysfunction facilitates neuronal hyperexcitability and may contribute to epilepsy. Genetic screens demonstrate that mutations of KCNJ10, the gene encoding Kir4.1, causes SeSAME/EAST syndrome, which is characterized by early onset seizures, compromised verbal and motor skills, profound cognitive deficits, and salt-wasting. KCNJ10 has also been linked to developmental disorders including autism. Cerebral trauma, ischemia, and inflammation are all associated with decreased astrocytic Kir4.1 current amplitude and astrocytic dysfunction. Additionally, neurodegenerative diseases such as Alzheimer disease and amyotrophic lateral sclerosis demonstrate loss of Kir4.1. This is particularly exciting in the context of Huntington disease, another neurodegenerative disorder in which restoration of Kir4.1 ameliorated motor deficits, decreased medium spiny neuron hyperexcitability, and extended survival in mouse models. Understanding the expression and regulation of Kir4.1 will be critical in determining if this channel can be exploited for therapeutic benefit.
Collapse
Affiliation(s)
- Sinifunanya E Nwaobi
- Department of Cell, Developmental and Integrative Biology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, AL, 35294, UK
| | - Vishnu A Cuddapah
- Department of Cell, Developmental and Integrative Biology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, AL, 35294, UK
| | - Kelsey C Patterson
- Department of Cell, Developmental and Integrative Biology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, AL, 35294, UK
| | - Anita C Randolph
- Department of Cell, Developmental and Integrative Biology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, AL, 35294, UK
| | - Michelle L Olsen
- Department of Cell, Developmental and Integrative Biology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, AL, 35294, UK.
| |
Collapse
|
22
|
Köller H, Fischer HG. Cytokines and Virus Proteins: Modulators of Glial Electrophysiological Properties. Neuroscientist 2016. [DOI: 10.1177/107385849900500310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cytokines are released during acute and chronic inflammatory diseases of the CNS and activate receptors on glial cells, thereby inducing various effects such as proliferation, expression of major histocompatibility complex genes or secretion of growth factors. Here, we summarize current evidence indicating that K+ currents, Ca 2+ currents, and the activity of ion transporters on astrocytes, microglial cells, and oligodendrocytes are also affected by cytokines. In disease states with associated elevated cytokine titers, such alterations in electrophysiological properties of glial cells might contribute to the patho genesis of neurological symptoms. NEUROSCIENTIST 5:142-146, 1999
Collapse
|
23
|
Moshrefi-Ravasdjani B, Dublin P, Seifert G, Jennissen K, Steinhäuser C, Kafitz KW, Rose CR. Changes in the proliferative capacity of NG2 cell subpopulations during postnatal development of the mouse hippocampus. Brain Struct Funct 2016; 222:831-847. [PMID: 27306788 DOI: 10.1007/s00429-016-1249-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/05/2016] [Indexed: 12/29/2022]
Abstract
Besides astrocytes and oligodendrocytes, NG2 proteoglycan-expressing cells (NG2 glia) represent a third subtype of macroglia in the brain. Originally described as oligodendrocyte precursor cells, they feature several characteristics not expected from mere progenitor cells, including synaptic connections with neurons. There is accumulating evidence that the properties of NG2 glia differ between different brain regions and developmental stages. To further analyze this proposed heterogeneity, we studied electrophysiological properties, transcript and protein expression, distribution and proliferative capacity of NG2 glia during postnatal development, focusing on the hippocampus and corpus callosum. All NG2 glia displayed a 'complex' current pattern consisting of voltage- and time-dependent in- and outward currents. In juvenile mice, Kir current densities and rectification index were highly variable and on average significantly lower than in adult animals. Single cell RT-PCR analyses of electrophysiologically characterized cells demonstrated that different glial genes were expressed at variable extent, independent of developmental stage and genetic background. In the hippocampus proper and the corpus callosum, the density of NG2 glia was highest at postnatal days (P)10-12, decreased by ~50 % at P25-35 and then remained stable in adults (P80-90). Interestingly, co-expression of NG2 and S100β, a marker for mature astrocytes, increased from 7 % at P10-12 to 27 % at P25-35 in the hippocampus proper, and then dropped again in the stratum radiatum at P80-90. In the dentate gyrus and corpus callosum, co-expression of NG2 and S100β was very low (3 %) and constant throughout development. Age-related differences were also observed with Ki-67, a proliferation marker. In NG2 glia of the CA1 region, its expression decreased from 16 % at P10-12 to 9 % (P25-35) and then 3 % (P80-90). Triple-stainings revealed that Ki-67 was also expressed in 2-3 % of NG2/S100β-positive cells in the juvenile and mature stratum radiatum, indicating that the latter, in contrast to S100β-positive astrocytes, still host proliferative potential. Taken together, we found significant differences in transcript and protein expression, electrophysiological properties and proliferative capacity of NG2 glia in the mouse forebrain, suggesting the co-existence of several subpopulations of NG2 glia. Our data thus support the idea of a substantial regional and developmental heterogeneity in this subtype of macroglia.
Collapse
Affiliation(s)
| | - Pavel Dublin
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Gerald Seifert
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53105, Bonn, Germany
| | - Katja Jennissen
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53105, Bonn, Germany
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53105, Bonn, Germany
| | - Karl W Kafitz
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Christine R Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany.
| |
Collapse
|
24
|
Zhong S, Du Y, Kiyoshi CM, Ma B, Alford CC, Wang Q, Yang Y, Liu X, Zhou M. Electrophysiological behavior of neonatal astrocytes in hippocampal stratum radiatum. Mol Brain 2016; 9:34. [PMID: 27004553 PMCID: PMC4802662 DOI: 10.1186/s13041-016-0213-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 03/12/2016] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Neonatal astrocytes are diverse in origin, and undergo dramatic change in gene expression, morphological differentiation and syncytial networking throughout development. Neonatal astrocytes also play multifaceted roles in neuronal circuitry establishment. However, the extent to which neonatal astrocytes differ from their counterparts in the adult brain remains unknown. RESULTS Based on ALDH1L1-eGFP expression or sulforhodamine 101 staining, neonatal astrocytes at postnatal day 1-3 can be reliably identified in hippocampal stratum radiatum. They exhibit a more negative resting membrane potential (V M), -85 mV, than mature astrocytes, -80 mV and a variably rectifying whole-cell current profile due to complex expression of voltage-gated outward transient K(+) (IKa), delayed rectifying K(+) (IKd) and inward K(+) (IKin) conductances. Differing from NG2 glia, depolarization-induced inward Na(+) currents (INa) could not be detected in neonatal astrocytes. A quasi-physiological V M of -69 mV was retained when inwardly rectifying Kir4.1 was inhibited by 100 μM Ba(2+) in both wild type and TWIK-1/TREK-1 double gene knockout astrocytes, indicating expression of additional leak K(+) channels yet unknown. In dual patch recording, electrical coupling was detected in 74 % (14/19 pairs) of neonatal astrocytes with largely variable coupling coefficients. The increasing gap junction coupling progressively masked the rectifying K(+) conductances to account for an increasing number of linear voltage-to-current relationship passive astrocytes (PAs). Gap junction inhibition, by 100 μM meclofenamic acid, substantially reduced membrane conductance and converted all the neonatal PAs to variably rectifying astrocytes. The low density expression of leak K(+) conductance in neonatal astrocytes corresponded to a ~50 % less K(+) uptake capacity compared to adult astrocytes. CONCLUSIONS Neonatal astrocytes predominantly express a variety of rectifying K(+) conductances, form discrete cell-to-cell gap junction coupling and are deficient in K(+) homeostatic capacity.
Collapse
Affiliation(s)
- Shiying Zhong
- />Department of Neurology, Shanghai 10th Hospital of Tongji University, School of Medicine, 301 Yan Chang Zhong Road, Shanghai, 200072 China
- />Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Yixing Du
- />Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Conrad M. Kiyoshi
- />Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Baofeng Ma
- />Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Catherine C. Alford
- />Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Qi Wang
- />Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Yongjie Yang
- />Department of Neuroscience, Tufts University School of Medicine, Boston, MA USA
| | - Xueyuan Liu
- />Department of Neurology, Shanghai 10th Hospital of Tongji University, School of Medicine, 301 Yan Chang Zhong Road, Shanghai, 200072 China
| | - Min Zhou
- />Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| |
Collapse
|
25
|
Astrocyte Cultures Mimicking Brain Astrocytes in Gene Expression, Signaling, Metabolism and K + Uptake and Showing Astrocytic Gene Expression Overlooked by Immunohistochemistry and In Situ Hybridization. Neurochem Res 2016; 42:254-271. [PMID: 26818759 DOI: 10.1007/s11064-016-1828-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/01/2016] [Accepted: 01/04/2016] [Indexed: 01/31/2023]
Abstract
Based on differences in gene expression between cultured astrocytes and freshly isolated brain astrocytes it has been claimed that cultured astrocytes poorly reflect the characteristics of their in vivo counterparts. This paper shows that this is not the case with the cultures of mouse astrocytes we have used since 1978. The culture is prepared following guidelines provided by Drs. Monique Sensenbrenner and John Booher, with the difference that dibutyryl cyclic AMP is added to the culture medium from the beginning of the third week. This addition has only minor effects on glucose and glutamate metabolism, but it is crucial for effects by elevated K+ concentrations and for Ca2+ homeostasis, important aspects of astrocyte function. Work by Liang Peng and her colleagues has shown identity between not only gene expression but also drug-induced gene upregulations and editings in astrocytes cultured by this method and astrocytes freshly isolated from brains of drug-treated animals. Dr. Norenberg's laboratory has demonstrated identical upregulation of the cotransporter NKCC1 in ammonia-exposed astrocytes and rats with liver failure. Similarity between cultured and freshly isolated astrocytes has also been shown in metabolism, K+ uptake and several aspects of signaling. However, others have shown that the gene for the glutamate transporter GLT1 is not expressed, and rat cultures show some abnormalities in K+ effects. Nevertheless, the overall reliability of the cultured cells is important because immunohistochemistry and in situ hybridization poorly demonstrate many astrocytic genes, e.g., those of nucleoside transporters, and even microarray analysis of isolated cells can be misleading.
Collapse
|
26
|
Platel JC, Bordey A. The multifaceted subventricular zone astrocyte: From a metabolic and pro-neurogenic role to acting as a neural stem cell. Neuroscience 2015; 323:20-8. [PMID: 26546469 DOI: 10.1016/j.neuroscience.2015.10.053] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 10/22/2015] [Accepted: 10/28/2015] [Indexed: 10/22/2022]
Abstract
A few decades ago it was discovered that two regions of the adult brain retain the ability to generate new neurons. These regions include the subgranular zone of the hippocampal dentate gyrus and the ventricular-subventricular zone (V-SVZ) located at the border of the lateral ventricle. In the V-SVZ, it was discovered that neural progenitor cells (NPCs) share many features of mature astrocytes and are often referred as V-SVZ astrocytes. We will first describe the markers, the morphology, and the neurophysiological characteristics of the mouse V-SVZ astrocytes. We will then discuss the fact that V-SVZ astrocytes constitute a mixed population with respect to their neurogenic properties, e.g., quiescent versus activated state, neurogenic fate, and transcription factors expression. Finally, we will describe two functions of V-SVZ astrocytes, their metabolic coupling to blood vessels and their neurogenic-supportive role consisting of providing guidance and survival cues to migrating newborn neurons.
Collapse
Affiliation(s)
- J C Platel
- Aix-Marseille University, Centre National de la Recherche Scientifique, Marseille, IBDM, UMR7288, Marseille, France.
| | - A Bordey
- Department of Neurosurgery and Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
27
|
Developmental expression of Kir4.1 in astrocytes and oligodendrocytes of rat somatosensory cortex and hippocampus. Int J Dev Neurosci 2015; 47:198-205. [DOI: 10.1016/j.ijdevneu.2015.09.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 09/16/2015] [Accepted: 09/16/2015] [Indexed: 12/31/2022] Open
|
28
|
Rela L, Piantanida AP, Bordey A, Greer CA. Voltage-dependent K+ currents contribute to heterogeneity of olfactory ensheathing cells. Glia 2015; 63:1646-59. [PMID: 25856239 DOI: 10.1002/glia.22834] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 03/24/2015] [Indexed: 02/03/2023]
Abstract
The olfactory nerve is permissive for axon growth throughout life. This has been attributed in part to the olfactory ensheathing glial cells that encompass the olfactory sensory neuron fascicles. Olfactory ensheathing cells (OECs) also promote axon growth in vitro and when transplanted in vivo to sites of injury. The mechanisms involved remain largely unidentified owing in part to the limited knowledge of the physiological properties of ensheathing cells. Glial cells rely for many functions on the properties of the potassium channels expressed; however, those expressed in ensheathing cells are unknown. Here we show that OECs express voltage-dependent potassium currents compatible with inward rectifier (Kir ) and delayed rectifier (KDR ) channels. Together with gap junction coupling, these contribute to the heterogeneity of membrane properties observed in OECs. The relevance of K(+) currents expressed by ensheathing cells is discussed in relation to plasticity of the olfactory nerve.
Collapse
Affiliation(s)
- Lorena Rela
- Departments of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut.,Systems Neuroscience Section, Department of Physiology and Biophysics, School of Medicine, University of Buenos Aires, Argentina.,Institute of Physiology and Biophysics Bernardo Houssay (IFIBIO Houssay-CONICET), Buenos Aires, Argentina
| | - Ana Paula Piantanida
- Systems Neuroscience Section, Department of Physiology and Biophysics, School of Medicine, University of Buenos Aires, Argentina.,Institute of Physiology and Biophysics Bernardo Houssay (IFIBIO Houssay-CONICET), Buenos Aires, Argentina
| | - Angelique Bordey
- Departments of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut.,Yale University School of Medicine, Departments of Cellular and Molecular Physiology, New Haven, Connecticut
| | - Charles A Greer
- Departments of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut.,Yale University School of Medicine, Departments of Neurobiology, New Haven, Connecticut
| |
Collapse
|
29
|
Sepúlveda FV, Pablo Cid L, Teulon J, Niemeyer MI. Molecular aspects of structure, gating, and physiology of pH-sensitive background K2P and Kir K+-transport channels. Physiol Rev 2015; 95:179-217. [PMID: 25540142 DOI: 10.1152/physrev.00016.2014] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
K(+) channels fulfill roles spanning from the control of excitability to the regulation of transepithelial transport. Here we review two groups of K(+) channels, pH-regulated K2P channels and the transport group of Kir channels. After considering advances in the molecular aspects of their gating based on structural and functional studies, we examine their participation in certain chosen physiological and pathophysiological scenarios. Crystal structures of K2P and Kir channels reveal rather unique features with important consequences for the gating mechanisms. Important tasks of these channels are discussed in kidney physiology and disease, K(+) homeostasis in the brain by Kir channel-equipped glia, and central functions in the hearing mechanism in the inner ear and in acid secretion by parietal cells in the stomach. K2P channels fulfill a crucial part in central chemoreception probably by virtue of their pH sensitivity and are central to adrenal secretion of aldosterone. Finally, some unorthodox behaviors of the selectivity filters of K2P channels might explain their normal and pathological functions. Although a great deal has been learned about structure, molecular details of gating, and physiological functions of K2P and Kir K(+)-transport channels, this has been only scratching at the surface. More molecular and animal studies are clearly needed to deepen our knowledge.
Collapse
Affiliation(s)
- Francisco V Sepúlveda
- Centro de Estudios Científicos, Valdivia, Chile; UPMC Université Paris 06, Team 3, Paris, France; and Institut National de la Santé et de la Recherche Médicale, UMR_S 1138, Paris, France
| | - L Pablo Cid
- Centro de Estudios Científicos, Valdivia, Chile; UPMC Université Paris 06, Team 3, Paris, France; and Institut National de la Santé et de la Recherche Médicale, UMR_S 1138, Paris, France
| | - Jacques Teulon
- Centro de Estudios Científicos, Valdivia, Chile; UPMC Université Paris 06, Team 3, Paris, France; and Institut National de la Santé et de la Recherche Médicale, UMR_S 1138, Paris, France
| | - María Isabel Niemeyer
- Centro de Estudios Científicos, Valdivia, Chile; UPMC Université Paris 06, Team 3, Paris, France; and Institut National de la Santé et de la Recherche Médicale, UMR_S 1138, Paris, France
| |
Collapse
|
30
|
Robel S, Buckingham SC, Boni JL, Campbell SL, Danbolt NC, Riedemann T, Sutor B, Sontheimer H. Reactive astrogliosis causes the development of spontaneous seizures. J Neurosci 2015; 35:3330-45. [PMID: 25716834 PMCID: PMC4339349 DOI: 10.1523/jneurosci.1574-14.2015] [Citation(s) in RCA: 195] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 12/16/2014] [Accepted: 01/12/2015] [Indexed: 11/21/2022] Open
Abstract
Epilepsy is one of the most common chronic neurologic diseases, yet approximately one-third of affected patients do not respond to anticonvulsive drugs that target neurons or neuronal circuits. Reactive astrocytes are commonly found in putative epileptic foci and have been hypothesized to be disease contributors because they lose essential homeostatic capabilities. However, since brain pathology induces astrocytes to become reactive, it is difficult to distinguish whether astrogliosis is a cause or a consequence of epileptogenesis. We now present a mouse model of genetically induced, widespread chronic astrogliosis after conditional deletion of β1-integrin (Itgβ1). In these mice, astrogliosis occurs in the absence of other pathologies and without BBB breach or significant inflammation. Electroencephalography with simultaneous video recording revealed that these mice develop spontaneous seizures during the first six postnatal weeks of life and brain slices show neuronal hyperexcitability. This was not observed in mice with neuronal-targeted β1-integrin deletion, supporting the hypothesis that astrogliosis is sufficient to induce epileptic seizures. Whole-cell patch-clamp recordings from astrocytes further suggest that the heightened excitability was associated with impaired astrocytic glutamate uptake. Moreover, the relative expression of the cation-chloride cotransporters (CCC) NKCC1 (Slc12a2) and KCC2 (Slc12a5), which are responsible for establishing the neuronal Cl(-) gradient that governs GABAergic inhibition were altered and the NKCC1 inhibitor bumetanide eliminated seizures in a subgroup of mice. These data suggest that a shift in the relative expression of neuronal NKCC1 and KCC2, similar to that observed in immature neurons during development, may contribute to astrogliosis-associated seizures.
Collapse
Affiliation(s)
- Stefanie Robel
- Department of Neurobiology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35209,
| | - Susan C Buckingham
- Department of Neurobiology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35209
| | - Jessica L Boni
- Department of Neurobiology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35209
| | - Susan L Campbell
- Department of Neurobiology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35209
| | - Niels C Danbolt
- Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway, and
| | - Therese Riedemann
- Institute of Physiology, Department of Physiological Genomics, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany
| | - Bernd Sutor
- Institute of Physiology, Department of Physiological Genomics, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany
| | - Harald Sontheimer
- Department of Neurobiology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35209
| |
Collapse
|
31
|
Caiazzo M, Giannelli S, Valente P, Lignani G, Carissimo A, Sessa A, Colasante G, Bartolomeo R, Massimino L, Ferroni S, Settembre C, Benfenati F, Broccoli V. Direct conversion of fibroblasts into functional astrocytes by defined transcription factors. Stem Cell Reports 2014; 4:25-36. [PMID: 25556566 PMCID: PMC4297873 DOI: 10.1016/j.stemcr.2014.12.002] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 11/28/2014] [Accepted: 12/01/2014] [Indexed: 01/04/2023] Open
Abstract
Direct cell reprogramming enables direct conversion of fibroblasts into functional neurons and oligodendrocytes using a minimal set of cell-lineage-specific transcription factors. This approach is rapid and simple, generating the cell types of interest in one step. However, it remains unknown whether this technology can be applied to convert fibroblasts into astrocytes, the third neural lineage. Astrocytes play crucial roles in neuronal homeostasis, and their dysfunctions contribute to the origin and progression of multiple human diseases. Herein, we carried out a screening using several transcription factors involved in defining the astroglial cell fate and identified NFIA, NFIB, and SOX9 to be sufficient to convert with high efficiency embryonic and postnatal mouse fibroblasts into astrocytes (iAstrocytes). We proved both by gene-expression profiling and functional tests that iAstrocytes are comparable to native brain astrocytes. This protocol can be then employed to generate functional iAstrocytes for a wide range of experimental applications. NFIA, NFIB, and SOX9 reprogram fibroblasts into induced astrocytes (iAstrocytes) iAstrocytes reprogramming induces a global change in gene-expression profiling iAstrocytes are functionally comparable to native astrocytes NFIA, NFIB, and SOX9 induce an astrocytic phenotype in human fibroblasts
Collapse
Affiliation(s)
- Massimiliano Caiazzo
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy.
| | - Serena Giannelli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Pierluigi Valente
- Section of Physiology, Department of Experimental Medicine, University of Genoa and National Institute of Neuroscience, 16132 Genoa, Italy
| | - Gabriele Lignani
- Department of Neuroscience and Brain Technologies, Italian Institute of Technology, 16132 Genoa, Italy
| | | | - Alessandro Sessa
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Gaia Colasante
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Rosa Bartolomeo
- Telethon Institute of Genetics and Medicine, Naples 80131, Italy; Dulbecco Telethon Institute
| | - Luca Massimino
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Stefano Ferroni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Carmine Settembre
- Telethon Institute of Genetics and Medicine, Naples 80131, Italy; Dulbecco Telethon Institute; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Medical Genetics, Department of Medical and Translational Science Unit, Federico II University, Via Pansini 5, 80131 Naples, Italy
| | - Fabio Benfenati
- Section of Physiology, Department of Experimental Medicine, University of Genoa and National Institute of Neuroscience, 16132 Genoa, Italy; Department of Neuroscience and Brain Technologies, Italian Institute of Technology, 16132 Genoa, Italy
| | - Vania Broccoli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy.
| |
Collapse
|
32
|
Schreiner AE, Durry S, Aida T, Stock MC, Rüther U, Tanaka K, Rose CR, Kafitz KW. Laminar and subcellular heterogeneity of GLAST and GLT-1 immunoreactivity in the developing postnatal mouse hippocampus. J Comp Neurol 2014; 522:204-24. [PMID: 23939750 DOI: 10.1002/cne.23450] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 05/23/2013] [Accepted: 08/02/2013] [Indexed: 11/06/2022]
Abstract
Astrocytes express two sodium-coupled transporters, glutamate-aspartate transporter (GLAST) and glutamate transporter-1 (GLT-1), which are essential for the maintenance of low extracellular glutamate levels. We performed a comparative analysis of the laminar and subcellular expression profile of GLAST and GLT-1 in the developing postnatal mouse hippocampus by using immunohistochemistry and western blotting and employing high-resolution fluorescence microscopy. Astrocytes were identified by costaining with glial fibrillary acidic protein (GFAP) or S100β. In CA1, the density of GFAP-positive cells and GFAP expression rose during the first 2 weeks after birth, paralleled by a steady increase in GLAST immunoreactivity and protein content. Upregulation of GLT-1 was completed only at postnatal days (P) P20-25 and was thus delayed by about 10 days. GLAST staining was highest along the stratum pyramidale and was especially prominent in astrocytes at P3-5. GLAST immunoreactivity indicated no preferential localization to a specific cellular compartment. GLT-1 exhibited a laminar expression pattern from P10-15 on, with the highest immunoreactivity in the stratum lacunosum-moleculare. At the cellular level, GLT-1 immunoreactivity did not entirely cover astrocyte somata and exhibited clusters at processes. In neonatal and juvenile animals, discrete clusters of GLT-1 were also detected at perivascular endfeet. From these results, we conclude there is a remarkable subcellular heterogeneity of GLAST and GLT-1 expression in the developing hippocampus. The clustering of GLT-1 at astrocyte endfeet indicates that it might serve a specialized functional role at the blood-brain barrier during formation of the hippocampal network.
Collapse
Affiliation(s)
- Alexandra E Schreiner
- Institute of Neurobiology, Heinrich Heine University Duesseldorf, 40225, Duesseldorf, Germany
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Passlick S, Trotter J, Seifert G, Steinhäuser C, Jabs R. The NG2 Protein Is Not Required for Glutamatergic Neuron-NG2 Cell Synaptic Signaling. Cereb Cortex 2014; 26:51-7. [PMID: 25100858 DOI: 10.1093/cercor/bhu171] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
NG2 glial cells (as from now NG2 cells) are unique in receiving synaptic input from neurons. However, the components regulating formation and maintenance of these neuron-glia synapses remain elusive. The transmembrane protein NG2 has been considered a potential mediator of synapse formation and alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) clustering, because it contains 2 extracellular Laminin G/Neurexin/Sex Hormone-Binding Globulin domains, which in neurons are crucial for formation of transsynaptic neuroligin-neurexin complexes. NG2 is connected via Glutamate Receptor-Interacting Protein with GluA2/3-containing AMPARs, thereby possibly mediating receptor clustering in glial postsynaptic density. To elucidate the role of NG2 in neuron-glia communication, we investigated glutamatergic synaptic transmission in juvenile and aged hippocampal NG2 cells of heterozygous and homozygous NG2 knockout mice. Neuron-NG2 cell synapses readily formed in the absence of NG2. Short-term plasticity, synaptic connectivity, postsynaptic AMPAR current kinetics, and density were not affected by NG2 deletion. During development, an NG2-independent acceleration of AMPAR current kinetics and decreased synaptic connectivity were observed. Our results indicate that the lack of NG2 does not interfere with genesis and basic properties of neuron-glia synapses. In addition, we demonstrate frequent expression of neuroligins 1-3 in juvenile and aged NG2 cells, suggesting a role of these molecules in synapse formation between NG2 glia and neurons.
Collapse
Affiliation(s)
- Stefan Passlick
- Institute of Cellular Neurosciences, University of Bonn, Bonn 53105, Germany
| | | | - Gerald Seifert
- Institute of Cellular Neurosciences, University of Bonn, Bonn 53105, Germany
| | | | - Ronald Jabs
- Institute of Cellular Neurosciences, University of Bonn, Bonn 53105, Germany
| |
Collapse
|
34
|
Nwaobi SE, Lin E, Peramsetty SR, Olsen ML. DNA methylation functions as a critical regulator of Kir4.1 expression during CNS development. Glia 2014; 62:411-27. [PMID: 24415225 DOI: 10.1002/glia.22613] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 10/29/2013] [Accepted: 11/21/2013] [Indexed: 12/22/2022]
Abstract
Kir4.1, a glial-specific K+ channel, is critical for normal CNS development. Studies using both global and glial-specific knockout of Kir4.1 reveal abnormal CNS development with the loss of the channel. Specifically, Kir4.1 knockout animals are characterized by ataxia, severe hypomyelination, and early postnatal death. Additionally, Kir4.1 has emerged as a key player in several CNS diseases. Notably, decreased Kir4.1 protein expression occurs in several human CNS pathologies including CNS ischemic injury, spinal cord injury, epilepsy, ALS, and Alzheimer's disease. Despite the emerging significance of Kir4.1 in normal and pathological conditions, its mechanisms of regulation are unknown. Here, we report the first epigenetic regulation of a K+ channel in the CNS. Robust developmental upregulation of Kir4.1 expression in rats is coincident with reductions in DNA methylation of the Kir4.1 gene, KCNJ10. Chromatin immunoprecipitation reveals a dynamic interaction between KCNJ10 and DNA methyltransferase 1 during development. Finally, demethylation of the KCNJ10 promoter is necessary for transcription. These findings indicate DNA methylation is a key regulator of Kir4.1 transcription. Given the essential role of Kir4.1 in normal CNS development, understanding the regulation of this K+ channel is critical to understanding normal glial biology.
Collapse
Affiliation(s)
- Sinifunanya E Nwaobi
- Department of Cell, Developmental and Integrative Biology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | | | | | | |
Collapse
|
35
|
Qian Z, Liu DJ, Liu Y, Han LM, Yuan M, Li JN, Xu B, Lu XL, Cao PX, Wang HY, Pan XD, Wang LJ, Qiao GF, Li BY. Increase in neuroexcitability of unmyelinated C-type vagal ganglion neurons during initial postnatal development of visceral afferent reflex functions. CNS Neurosci Ther 2013; 19:954-62. [PMID: 24164691 DOI: 10.1111/cns.12195] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 09/22/2013] [Accepted: 09/23/2013] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Baroreflex gain increase up closely to adult level during initial postnatal weeks, and any interruption within this period will increase the risk of cardiovascular problems in later of life span. We hypothesize that this short period after birth might be critical for postnatal development of vagal ganglion neurons (VGNs). METHODS To evaluate neuroexcitability evidenced by discharge profiles and coordinate changes, ion currents were collected from identified A- and C-type VGNs at different developmental stages using whole-cell patch clamping. RESULTS C-type VGNs underwent significant age-dependent transition from single action potential (AP) to repetitive discharge. The coordinate changes between TTX-S and TTX-R Na(+) currents were also confirmed and well simulated by computer modeling. Although 4-AP or iberiotoxin age dependently increased firing frequency, AP duration was prolonged in an opposite fashion, which paralleled well with postnatal changes in 4-AP- and iberiotoxin-sensitive K(+) current activity, whereas less developmental changes were verified in A-types. CONCLUSION These data demonstrate for the first time that the neuroexcitability of C-type VGNs increases significantly compared with A-types within initial postnatal weeks evidenced by AP discharge profiles and coordinate ion channel changes, which explain, at least in part, that initial postnatal weeks may be crucial for ontogenesis in visceral afferent reflex function.
Collapse
Affiliation(s)
- Zhao Qian
- Department of Pharmacology, Harbin Medical University, Harbin, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Dallérac G, Chever O, Rouach N. How do astrocytes shape synaptic transmission? Insights from electrophysiology. Front Cell Neurosci 2013; 7:159. [PMID: 24101894 PMCID: PMC3787198 DOI: 10.3389/fncel.2013.00159] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 09/02/2013] [Indexed: 02/01/2023] Open
Abstract
A major breakthrough in neuroscience has been the realization in the last decades that the dogmatic view of astroglial cells as being merely fostering and buffering elements of the nervous system is simplistic. A wealth of investigations now shows that astrocytes actually participate in the control of synaptic transmission in an active manner. This was first hinted by the intimate contacts glial processes make with neurons, particularly at the synaptic level, and evidenced using electrophysiological and calcium imaging techniques. Calcium imaging has provided critical evidence demonstrating that astrocytic regulation of synaptic efficacy is not a passive phenomenon. However, given that cellular activation is not only represented by calcium signaling, it is also crucial to assess concomitant mechanisms. We and others have used electrophysiological techniques to simultaneously record neuronal and astrocytic activity, thus enabling the study of multiple ionic currents and in depth investigation of neuro-glial dialogues. In the current review, we focus on the input such approach has provided in the understanding of astrocyte-neuron interactions underlying control of synaptic efficacy.
Collapse
Affiliation(s)
- Glenn Dallérac
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, CNRS UMR 7241, INSERM U1050, Collège de France Paris, France
| | | | | |
Collapse
|
37
|
Abstract
Spermine (SPM) and spermidine, endogenous polyamines with the ability to modulate various ion channels and receptors in the brain, exert neuroprotective, antidepressant, antioxidant, and other effects in vivo such as increasing longevity. These polyamines are preferably accumulated in astrocytes, and we hypothesized that SPM increases glial intercellular communication by interacting with glial gap junctions. The results obtained in situ, using Lucifer yellow propagation in the astrocytic syncitium of 21-25-day-old rat CA1 hippocampal slices, showed reduced coupling when astrocytes were dialyzed with standard intracellular solutions without SPM. However, there was a robust increase in the spreading of Lucifer yellow through gap junctions to neighboring astrocytes when the cells were patched with intracellular solutions containing 1 mM SPM, a physiological concentration in glia. Lucifer yellow propagation was inhibited by gap junction blockers. Our findings show that the glial syncitium propagates SPM through gap junctions and further indicate a new role of polyamines in the regulation of the astroglial network under both normal and pathological conditions.
Collapse
|
38
|
Sun L, Yu Z, Wang W, Liu X. Both NKCC1 and anion exchangers contribute to Cl⁻ accumulation in postnatal forebrain neuronal progenitors. Eur J Neurosci 2012; 35:661-72. [PMID: 22390178 DOI: 10.1111/j.1460-9568.2012.08007.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Neuronal progenitors are continuously generated in the postnatal rodent subventricular zone and migrate along the rostral migratory stream to supply interneurons in the olfactory bulb. Nonsynaptic GABAergic signaling affects the postnatal neurogenesis by depolarizing neuronal progenitors, which depends on an elevated intracellular Cl(-) concentration. However, the molecular mechanism responsible for Cl(-) accumulation in these cells still remains elusive. Using confocal Ca(2+) imaging, we found that GABA depolarization-induced Ca(2+) increase was either abolished by bumetanide, a specific inhibitor of the Na(+) -K(+) -2Cl(-) cotransporter, or reduced by partial replacement of extracellular Na(+) with Li(+) , in the HEPES buffer but not in the CO(2)/HCO₃⁻ buffer. GABA depolarization-induced Ca(2+) increase in CO(2)/HCO₃⁻ buffer was abolished by a combination of bumetanide with the anion exchanger inhibitor DIDS or with the carbonic anhydrase inhibitor acetozalimide. Using gramicidin-perforated patch-clamp recording, we further confirmed that bumetanide, together with DIDS or acetozalimide, reduced the intracellular chloride concentration in the neuronal progenitors. In addition, with BrdU labeling, we demonstrated that blocking of the Na(+) -K(+) -2Cl(-) cotransporter, but not anion exchangers, reduced the proliferation of neuronal progenitors. Our results indicate that both the Na(+) -K(+) -2Cl(-) cotransporter and anion exchangers contribute to the elevated intracellular chloride responsible for the depolarizing action of GABA in the postnatal forebrain neuronal progenitors. However, the Na(+) -K(+) -2Cl(-) cotransporter displays an additional effect on neuronal progenitor proliferation.
Collapse
Affiliation(s)
- Lin Sun
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT 06520-8001, USA
| | | | | | | |
Collapse
|
39
|
Braganza O, Bedner P, Hüttmann K, von Staden E, Friedman A, Seifert G, Steinhäuser C. Albumin is taken up by hippocampal NG2 cells and astrocytes and decreases gap junction coupling. Epilepsia 2012; 53:1898-906. [PMID: 22967085 DOI: 10.1111/j.1528-1167.2012.03665.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Dysfunction of the blood-brain barrier (BBB) and albumin extravasation have been suggested to play a role in the etiology of human epilepsy. In this context, dysfunction of glial cells attracts increasing attention. Our study was aimed to analyze in the hippocampus (1) which cell types internalize albumin injected into the lateral ventricle in vivo, (2) whether internalization into astrocytes impacts their coupling and expression of connexin 43 (Cx43), and (3) whether expression of Kir4.1, the predominating astrocytic K(+) channel subunit, is altered by albumin. METHODS The patch-clamp method was combined with single cell tracer filling to investigate electrophysiologic properties and gap junction coupling (GJC). For cell identification, mice with cell type-specific expression of a fluorescent protein (NG2kiEYFP mice) and immunohistochemistry were employed. Semiquantitative real time polymerase chain reaction (RT-PCR) allowed analysis of Kir4.1 and Cx43 transcript levels. KEY FINDINGS We show that fluorescently labeled albumin is taken up by astrocytes, NG2 cells, and neurons, with NG2 cells standing out in terms of the quantity of uptake. Within 5 days postinjection (dpi), intracellular albumin accumulation was largely reduced suggesting rapid degradation. Electrophysiologic analysis of astrocytes and NG2 cells revealed no changes in their membrane properties at either time point. However, astrocytic GJC was significantly decreased at 1 dpi but returned to control levels within 5 dpi. We found no changes in hippocampal Cx43 transcript expression, suggesting that other mechanisms account for the observed changes in coupling. Kir4.1 mRNA was regulated oppositely in the CA1 stratum radiatum, with a strong increase at 1 dpi followed by a decrease at 5 dpi. SIGNIFICANCE The present study demonstrates that extravasal albumin in the hippocampus induces rapid changes of astrocyte function, which can be expected to impair ion and transmitter homeostasis and contribute to hyperactivity and epileptogenesis. Therefore, astrocytes may represent alternative targets for antiepileptic therapeutic approaches.
Collapse
Affiliation(s)
- Oliver Braganza
- Institute of Cellular Neurosciences, University of Bonn, Sigmund Freud Strasse 25, Bonn, Germany
| | | | | | | | | | | | | |
Collapse
|
40
|
Karpuk N, Burkovetskaya M, Kielian T. Neuroinflammation alters voltage-dependent conductance in striatal astrocytes. J Neurophysiol 2012; 108:112-23. [PMID: 22457466 DOI: 10.1152/jn.01182.2011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neuroinflammation has the capacity to alter normal central nervous system (CNS) homeostasis and function. The objective of the present study was to examine the effects of an inflammatory milieu on the electrophysiological properties of striatal astrocyte subpopulations with a mouse bacterial brain abscess model. Whole cell patch-clamp recordings were performed in striatal glial fibrillary acidic protein (GFAP)-green fluorescent protein (GFP)(+) astrocytes neighboring abscesses at postinfection days 3 or 7 in adult mice. Cell input conductance (G(i)) measurements spanning a membrane potential (V(m)) surrounding resting membrane potential (RMP) revealed two prevalent astrocyte subsets. A1 and A2 astrocytes were identified by negative and positive G(i) increments vs. V(m), respectively. A1 and A2 astrocytes displayed significantly different RMP, G(i), and cell membrane capacitance that were influenced by both time after bacterial exposure and astrocyte proximity to the inflammatory site. Specifically, the percentage of A1 astrocytes was decreased immediately surrounding the inflammatory lesion, whereas A2 cells were increased. These changes were particularly evident at postinfection day 7, revealing increased cell numbers with an outward current component. Furthermore, RMP was inversely modified in A1 and A2 astrocytes during neuroinflammation, and resting G(i) was increased from 21 to 30 nS in the latter. In contrast, gap junction communication was significantly decreased in all astrocyte populations associated with inflamed tissues. Collectively, these findings demonstrate the heterogeneity of striatal astrocyte populations, which experience distinct electrophysiological modifications in response to CNS inflammation.
Collapse
Affiliation(s)
- Nikolay Karpuk
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5900, USA
| | | | | |
Collapse
|
41
|
Béchade C, Pascual O, Triller A, Bessis A. Nitric oxide regulates astrocyte maturation in the hippocampus: involvement of NOS2. Mol Cell Neurosci 2011; 46:762-9. [PMID: 21354308 DOI: 10.1016/j.mcn.2011.02.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 02/16/2011] [Accepted: 02/18/2011] [Indexed: 11/27/2022] Open
Abstract
Neurons and astrocytes are generated sequentially from radial glia. Once neurogenesis is completed, radial glia starts to differentiate into immature astrocytes. Astrocytes then maturate and change their morphology and electrophysiological properties. Neurotrophic cytokines or bone morphogenetic proteins have been identified as inducers of the developmental switch from neurogenesis to astrogenesis. However, the factors and mechanisms regulating the late differentiation of radial glia and the subsequent astrocyte maturation are poorly described. We used two independent approaches to examine the role of nitric oxide (NO) in the process of astrogenesis and maturation of astrocytes. First using a pharmacological approach, we depleted NO from developing hippocampus by intraventricular injection of a specific scavenger. Then by a genetic approach, we analyzed a nitric oxide synthase-2 (NOS2) knockout mouse. In both models, we found that differentiation of RC2-positive radial glia into late GFAP-positive radial glia was impaired. The cell-fate analysis after incorporation of BrdU demonstrated that astrogenesis was not altered upon NOS2 deficiency. Maturation of astrocytes was assessed by electrophysiological recordings at P7 and functional analysis. In wild type, 20% of astrocytes were immature as shown by their non-linear I/V relationship and high membrane resistance, whereas in NOS2-/- hippocampus, 51% of the astrocytes displayed an immature profile. The reduced branching of astrocytes upon NOS2 deficiency and their low content in connexin-43 further confirmed their immature profile. Our results highlight a novel developmental role of NO and NOS2 in the differentiation of radial glia and the maturation of astrocytes.
Collapse
Affiliation(s)
- Catherine Béchade
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Inserm U1024, Paris F-75005, France
| | | | | | | |
Collapse
|
42
|
Montiel-Herrera M, García-Colunga J. Current profiles of astrocytes from the corpus callosum of newborn and 28-day-old rats. Neurosci Lett 2010; 485:189-93. [DOI: 10.1016/j.neulet.2010.09.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Revised: 08/06/2010] [Accepted: 09/02/2010] [Indexed: 11/16/2022]
|
43
|
Lin YC, Liu YC, Huang YY, Lien CC. High-density expression of Ca2+-permeable ASIC1a channels in NG2 glia of rat hippocampus. PLoS One 2010; 5:e12665. [PMID: 20844750 PMCID: PMC2937019 DOI: 10.1371/journal.pone.0012665] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Accepted: 08/16/2010] [Indexed: 01/25/2023] Open
Abstract
NG2 cells, a fourth type of glial cell in the mammalian CNS, undergo reactive changes in response to a wide variety of brain insults. Recent studies have demonstrated that neuronally expressed acid-sensing ion channels (ASICs) are implicated in various neurological disorders including brain ischemia and seizures. Acidosis is a common feature of acute neurological conditions. It is postulated that a drop in pH may be the link between the pathological process and activation of NG2 cells. Such postulate immediately prompts the following questions: Do NG2 cells express ASICs? If so, what are their functional properties and subunit composition? Here, using a combination of electrophysiology, Ca2+ imaging and immunocytochemistry, we present evidence to demonstrate that NG2 cells of the rat hippocampus express high density of Ca2+-permeable ASIC1a channels compared with several types of hippocampal neurons. First, nucleated patch recordings from NG2 cells revealed high density of proton-activated currents. The magnitude of proton-activated current was pH dependent, with a pH for half-maximal activation of 6.3. Second, the current-voltage relationship showed a reversal close to the equilibrium potential for Na+. Third, psalmotoxin 1, a blocker specific for the ASIC1a channel, largely inhibited proton-activated currents. Fourth, Ca2+ imaging showed that activation of proton-activated channels led to an increase of [Ca2+]i. Finally, immunocytochemistry showed co-localization of ASIC1a and NG2 proteins in the hippocampus. Thus the acid chemosensor, the ASIC1a channel, may serve for inducing membrane depolarization and Ca2+ influx, thereby playing a crucial role in the NG2 cell response to injury following ischemia.
Collapse
Affiliation(s)
- Yen-Chu Lin
- Institute of Neuroscience and Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Chao Liu
- Institute of Neuroscience and Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Yin Huang
- Department of Anesthesiology, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Cheng-Chang Lien
- Institute of Neuroscience and Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
44
|
Integrins and ion channels in cell migration: implications for neuronal development, wound healing and metastatic spread. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 674:107-23. [PMID: 20549944 DOI: 10.1007/978-1-4419-6066-5_10] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cells migration is necessary for proper embryonic development and adult tissue remodeling. Its mechanisms determine the physiopathology of processes such as neuronal targeting, inflammation, wound healing and metastatic spread. Crawling of cells onto solid surfaces requires a controlled sequence of cell protrusions and retractions that mainly depends on sophisticated regulation of the actin cytoskeleton, although the contribution of microtubules should not be neglected. This process is triggered and modulated by a combination of diffusible and fixed environmental signals. External cues are sensed and integrated by membrane receptors, including integrins, which transduce these signals into cellular signaling pathways, often centered on the small GTPase proteins belonging to the Rho family. These pathways regulate the coordinated cytoskeletal rearrangements necessary for proper timing of adhesion, contraction and detachement at the front and rear side of cells finding their way through the extracellular spaces. The overall process involves continuous modulation of cell motility, shape and volume, in which ion channels play major roles. In particular, Ca2+ signals have both global and local regulatory effects on cell motility, because they target the contractile proteins as well as many regulatory proteins. After reviewing the fundamental mechanisms of eukaryotic cell migration onto solid substrates, we briefly describe how integrin receptors and ion channels are involved in cell movement. We next examine a few processes in which these mechanisms have been studied in depth. We thus illustrate how integrins and K+ channels control cell volume and migration, how intracellular Ca2+ homeostasis affects the motility of neuronal growth cones and what is known about the ion channel roles in epithelial cell migration. These mechanisms are implicated in a variety of pathological processes, such as the disruption of neural circuits and wound healing. Finally, we describe the interaction between neoplastic cells and their local environment and how derangement of adhesion can lead to metastatic spread. It is likely that the cellular mechanisms controlled by integrin receptors, ion channels or both participate in the entire metastatic process. Until now, however, evidence is limited to a few steps of the metastatic cascade, such as brain tumor invasiveness.
Collapse
|
45
|
Bergles DE, Jabs R, Steinhäuser C. Neuron-glia synapses in the brain. BRAIN RESEARCH REVIEWS 2010; 63:130-7. [PMID: 20018210 PMCID: PMC2862892 DOI: 10.1016/j.brainresrev.2009.12.003] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Revised: 12/04/2009] [Accepted: 12/08/2009] [Indexed: 11/19/2022]
Abstract
The ability to investigate the electrophysiological properties of individual cells in acute brain tissue led to the discovery that many glial cells have the capacity to respond rapidly to neuronal activity. In particular, a distinct class of neuroglial cells known as NG2 cells, which exhibit many of the properties that have been described for glial subtypes such as complex cells, polydendrocytes, synantocytes and GluR cells, express ionotropic receptors for glutamate and GABA. In both gray and white matter, NG2 cells form direct synaptic junctions with axons, which enable transient activation of these receptors. Electrophysiological analyses have shown that these neuron-glia synapses exhibit all the hallmarks of 'classical' neuron-neuron synapses, including rapid activation, quantized responses, facilitation and depression, and presynaptic inhibition. Electron microscopy indicates that axons form morphologically distinct junctions at discrete sites along processes of NG2 cells, suggesting that NG2 cells are an overt target of axonal projections. AMPA receptors expressed by NG2 cells exhibit varying degrees of Ca(2+) permeability, depending on the brain region and stage of development, and in white matter NG2 cells have also been shown to express functional NMDA receptors. Ca(2+) influx through AMPA receptors following repetitive stimulation can trigger long term potentiation of synaptic currents in NG2 cells. The expression of receptors with significant Ca(2+) permeability may increase the susceptibility of NG2 cells to excitotoxic injury. Future studies using transgenic mice in which expression of receptors can be manipulated selectively in NG2 cells have to define the functions of this enigmatic neuron-glia signaling in the normal and diseased CNS.
Collapse
Affiliation(s)
- Dwight E. Bergles
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Ronald Jabs
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Sigmund Freud Str. 25, 53105 Bonn, Germany
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Sigmund Freud Str. 25, 53105 Bonn, Germany
| |
Collapse
|
46
|
Costa MR, Götz M, Berninger B. What determines neurogenic competence in glia? ACTA ACUST UNITED AC 2010; 63:47-59. [DOI: 10.1016/j.brainresrev.2010.01.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Revised: 01/12/2010] [Accepted: 01/14/2010] [Indexed: 01/20/2023]
|
47
|
Langer J, Rose CR. Synaptically induced sodium signals in hippocampal astrocytes in situ. J Physiol 2010; 587:5859-77. [PMID: 19858225 DOI: 10.1113/jphysiol.2009.182279] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Astrocytes are in close contact to excitatory synapses and express transporters which mediate the sodium-dependent uptake of glutamate. In cultured astrocytes, selective activation of glutamate transport results in sodium elevations which stimulate Na(+)/K(+)-ATPase and glucose uptake, indicating that synaptic release of glutamate might couple excitatory neuronal activity to glial sodium homeostasis and metabolism. Here, we analysed intracellular sodium transients evoked by synaptic stimulation in acute mouse hippocampal slices using quantitative sodium imaging with the sodium-sensitive fluorescent indicator dye SBFI (sodium-binding benzofuran isophthalate). We found that short bursts of Schaffer collateral stimulation evoke sodium transients in the millimolar range in both CA1 pyramidal neurons and in SR101-positive astrocytes of the stratum radiatum. At low stimulation intensities, glial sodium transients were confined to one to two primary branches and adjacent fine processes and only weakly invaded the soma. Increasing the number of activated afferent fibres by increasing the stimulation intensity elicited global sodium transients detectable in the processes as well as the somata of astrocytes. Pharmacological analysis revealed that neuronal sodium signals were mainly attributable to sodium influx through ionotropic glutamate receptors. Activation of ionotropic receptors also contributed to glial sodium transients, while TBOA-sensitive glutamate transport was the major pathway responsible for sodium influx into astrocytes. Our results thus establish that glutamatergic synaptic transmission in the hippocampus results in sodium transients in astrocytes that are mainly mediated by activation of glutamate transport. They support the proposed link between excitatory synaptic activity, glutamate uptake and sodium signals in astrocytes of the hippocampus.
Collapse
Affiliation(s)
- Julia Langer
- Institute for Neurobiology, Heinrich-Heine-University Duesseldorf, Universitaetsstrasse 1, 26.02.00, 40225 Duesseldorf, Germany
| | | |
Collapse
|
48
|
Erecinska M, Cherian S, A Silver I. Brain development and susceptibility to damage; ion levels and movements. Curr Top Dev Biol 2009; 69:139-86. [PMID: 16243599 DOI: 10.1016/s0070-2153(05)69006-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Responses of immature brains to physiological and pathological stimuli often differ from those in the adult. Because CNS function critically depends on ion movements, this chapter evaluates ion levels and gradients during ontogeny and their alterations in response to adverse conditions. Total brain Na(+) and Cl(-) content decreases during development, but K(+) content rises, reflecting shrinkage of the extracellular and increase in the intracellular water spaces and a reduction in total brain water volume. Unexpectedly, [K(+)](i) seems to fall during the first postnatal week, which should reduce [K(+)](i)/ [K(+)](e) and result in a lower V(m), consistent with experimental observations. Neuronal [Cl(-)](i) is high during early postnatal development, hence the opening of Cl(-) conduction pathways may lead to plasma membrane depolarization. Equivalent loss of K(+)(i) into a relatively large extracellular space leads to a smaller increase in [K(+)](e) in immature animals, while the larger reservoir of Ca(2+)(e) may result in a greater [Ca(2+)](i) rise. In vivo and in vitro studies show that compared with adult, developing brains are more resistant to hypoxic/ischemic ion leakage: increases in [K(+)](e) and decreases in [Ca(2+)](e) are slower and smaller, consistent with the known low level of energy utilization and better maintenance of [ATP]. Severe hypoxia/ischemia may, however, lead to large Ca(2+)(i) overload. Rises in [K(+)](e) during epileptogenesis in vivo are smaller and take longer to manifest themselves in immature brains, although the rate of K(+) clearance is slower. By contrast, in vitro studies suggest the existence of a period of enhanced vulnerability sometime during the developmental period. This chapter concludes that there is a great need for more information on ion changes during ontogeny and poses the question whether the rat is the most appropriate model for investigation of mechanisms of pathological changes in human neonates.
Collapse
Affiliation(s)
- Maria Erecinska
- Department of Anatomy, School of Veterinary Science, Bristol, United Kingdom
| | | | | |
Collapse
|
49
|
Sundelacruz S, Levin M, Kaplan DL. Role of membrane potential in the regulation of cell proliferation and differentiation. Stem Cell Rev Rep 2009; 5:231-46. [PMID: 19562527 PMCID: PMC10467564 DOI: 10.1007/s12015-009-9080-2] [Citation(s) in RCA: 329] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Accepted: 06/07/2009] [Indexed: 12/11/2022]
Abstract
Biophysical signaling, an integral regulator of long-term cell behavior in both excitable and non-excitable cell types, offers enormous potential for modulation of important cell functions. Of particular interest to current regenerative medicine efforts, we review several examples that support the functional role of transmembrane potential (V(mem)) in the regulation of proliferation and differentiation. Interestingly, distinct V(mem) controls are found in many cancer cell and precursor cell systems, which are known for their proliferative and differentiation capacities, respectively. Collectively, the data demonstrate that bioelectric properties can serve as markers for cell characterization and can control cell mitotic activity, cell cycle progression, and differentiation. The ability to control cell functions by modulating bioelectric properties such as V(mem) would be an invaluable tool for directing stem cell behavior toward therapeutic goals. Biophysical properties of stem cells have only recently begun to be studied and are thus in need of further characterization. Understanding the molecular and mechanistic basis of biophysical regulation will point the way toward novel ways to rationally direct cell functions, allowing us to capitalize upon the potential of biophysical signaling for regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Sarah Sundelacruz
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA 02155, USA
| | | | | |
Collapse
|
50
|
Vignali M, Benfenati V, Caprini M, Anderova M, Nobile M, Ferroni S. The endocannabinoid anandamide inhibits potassium conductance in rat cortical astrocytes. Glia 2009; 57:791-806. [PMID: 19031444 DOI: 10.1002/glia.20807] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Endocannabinoids are a family of endogenous signaling molecules that modulate neuronal excitability in the central nervous system (CNS) by interacting with cannabinoid (CB) receptors. In spite of the evidence that astroglial cells also possess CB receptors, there is no information on the role of endocannabinoids in regulating CNS function through the modulation of ion channel-mediated homeostatic mechanisms in astroglial cells. We provide electrophysiological evidence that the two brain endocannabinoids anandamide (AEA) and 2-arachidonylglycerol (2-AG) markedly depress outward conductance mediated by delayed outward rectifier potassium current (IK(DR)) in primary cultured rat cortical astrocytes. Pharmacological experiments suggest that the effect of AEA does not result from the activation of known CB receptors. Moreover, neither the production of AEA metabolites nor variations in free cytosolic calcium are involved in the negative modulation of IK(DR). We show that the action of AEA is mediated by its interaction with the extracellular leaflet of the plasma membrane. Similar experiments performed in situ in cortical slices indicate that AEA downregulates IK(DR) in complex and passive astroglial cells. Moreover, IK(DR) is also inhibited by AEA in NG2 glia. Collectively, these results support the notion that endocannabinoids may exert their modulation of CNS function via the regulation of homeostatic function of the astroglial syncytium mediated by ion channel activity.
Collapse
Affiliation(s)
- M Vignali
- Department of Human and General Physiology, University of Bologna, 40127 Bologna, Italy
| | | | | | | | | | | |
Collapse
|