Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cells ; 10(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33466322

ABSTRACT

Astrocytes contribute to many higher brain functions. A key mechanism in glia-to-neuron signalling is vesicular exocytosis; however, the identity of exocytosis organelles remains a matter of debate. Since vesicles derived from the trans-Golgi network (TGN) are not considered in this context, we studied the astrocyte TGN by immunocytochemistry applying anti-Rab6A. In mouse brain, Rab6A immunostaining is found to be unexpectedly massive, diffuse in all regions, and is detected preferentially and abundantly in the peripheral astrocyte processes, which is hardly evident without glial fibrillary acid protein (GFAP) co-staining. All cells positive for the astrocytic markers glutamine synthetase (GS), GFAP, aldehyde dehydrogenase 1 family member L1 (Aldh1L1), or SRY (sex determining region Y)-box 9 (SOX9) were Rab6A+. Rab6A is excluded from microglia, oligodendrocytes, and NG2 cells using cell type-specific markers. In human cortex, Rab6A labelling is very similar and associated with GFAP+ astrocytes. The mouse data also confirm the specific astrocytic labelling by Aldh1L1 or SOX9; the astrocyte-specific labelling by GS sometimes debated is replicated again. In mouse and human brain, individual astrocytes display high variability in Rab6A+ structures, suggesting dynamic regulation of the glial TGN. In summary, Rab6A expression is an additional, global descriptor of astrocyte identity. Rab6A might constitute an organelle system with a potential role of Rab6A in neuropathological and physiological processes.


Subject(s)
Antigens, Differentiation/metabolism , Astrocytes/metabolism , Cerebral Cortex/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Cell Line , Humans , Mice , Species Specificity
2.
Int J Mol Sci ; 20(15)2019 Aug 02.
Article in English | MEDLINE | ID: mdl-31382374

ABSTRACT

Astrocytes are increasingly perceived as active partners in physiological brain function and behaviour. The structural correlations of the glia-synaptic interaction are the peripheral astrocyte processes (PAPs), where ezrin and radixin, the two astrocytic members of the ezrin-radixin-moesin (ERM) family of proteins are preferentially localised. While the molecular mechanisms of ERM (in)activation appear universal, at least in mammalian cells, and have been studied in great detail, the actual ezrin and radixin kinases, phosphatases and binding partners appear cell type specific and may be multiplexed within a cell. In astrocytes, ezrin is involved in process motility, which can be stimulated by the neurotransmitter glutamate, through activation of the glial metabotropic glutamate receptors (mGluRs) 3 or 5. However, it has remained open how this mGluR stimulus is transduced to ezrin activation. Knowing upstream signals of ezrin activation, ezrin kinase(s), and membrane-bound binding partners of ezrin in astrocytes might open new approaches to the glial role in brain function. Ezrin has also been implicated in invasive behaviour of astrocytomas, and glial activation. Here, we review data pertaining to potential molecular interaction partners of ezrin in astrocytes, with a focus on PKC and GRK2, and in gliomas and other diseases, to stimulate further research on their potential roles in glia-synaptic physiology and pathology.


Subject(s)
Astrocytes/metabolism , Cytoskeletal Proteins/metabolism , Membrane Proteins/metabolism , Protein Interaction Maps , Animals , Astrocytes/pathology , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/pathology , Cytoskeletal Proteins/analysis , G-Protein-Coupled Receptor Kinase 2/analysis , G-Protein-Coupled Receptor Kinase 2/metabolism , Glioma/metabolism , Glioma/pathology , Humans , Membrane Proteins/analysis , Protein Kinase C/analysis , Protein Kinase C/metabolism
3.
J Neurosci Methods ; 320: 87-97, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30876913

ABSTRACT

BACKGROUND: In the analysis of animal models of CNS diseases such as experimental autoimmune encephalomyelitis (EAE), immunostaining and histopathology are important readouts. However, the complex morphological features of a tissue staining are often reduced to a single measure which relies on tedious manual planimetry. Furthermore, the measure itself and co-variables such as the region being analysed are chosen in a human decision-making process, which introduces bias. NEW METHOD: First aim of the present study is to provide an open-source workflow for the high-throughput, unsupervised quantification of different stainings in the spinal cord. We evaluate different EAE models, spinal cord regions and different time points of disease. By applying random forest classification, we compare different measures. RESULTS: Exemplified for glial reactivity, we show that measures and variables interact and that their values are non-normally distributed, hampering the common use of parametric tests. Furthermore, we demonstrate that one-dimensional measures are insufficient descriptors for immunofluorescence data in EAE and thus need to be considered as partly invalid. COMPARISON WITH EXISTING METHODS: We show in a systematic analysis of EAE studies that currently published immunohistological outcomes are highly incompatible regarding methodology and statistics. Furthermore, they lack the report of important information necessary for reproducibility and do not use unsupervised automatic analysis. CONCLUSIONS: Our results discover relevant caveats in the currently used methods of immunofluorescence analysis. The provided step-by-step instructions and open-source code are intended to serve as a framework for sensitive, unbiased immunofluorescence analysis of tissue sections in translational research.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Fluorescent Antibody Technique/methods , Image Processing, Computer-Assisted/methods , Neurosciences/methods , Spinal Cord/pathology , Animals , Fluorescent Antibody Technique/standards , Image Processing, Computer-Assisted/standards , Neurosciences/standards
4.
Methods Mol Biol ; 1938: 169-186, 2019.
Article in English | MEDLINE | ID: mdl-30617980

ABSTRACT

Assessing the amount and subcellular distribution of protein expression is a key component in modern cell biological and medical research. We studied protein kinase Cδ (PKCδ) as a potential regulator of mitochondrial metabolism in astrocytes, and sought to evaluate mitochondrial translocation of PKCδ since this is an important determinant of its function. Apart from visualizing compartment specific localization of mobile proteins such as PKCδ, we also wanted to determine what amount of a cell's total content of a particular protein is located to a specific compartment, or translocated comparing control and experimental condition.We develop a semiquantitative parameter that indicates the relative protein distribution to two subcellular compartments, starting from standard two-channel fluorescence microscopy images. We studied the mitochondrial translocation of PKCδ in astrocytes using double immunofluorescence microscopy and object-oriented image analysis. In one channel, the protein of interest (PKCδ) is labeled, in the other the compartment or organelle of interest (mitochondria, using cytochrome oxidase IV). Both channels were binarized, turned into object populations, and the channel specific values for total area and integrated intensity extracted. From these values, the "intensity density ratio" (IDR) is calculated, a standardized parameter to easily compare distribution patterns in different cells or ROIs. IDR is highly sensitive to changes in localization pattern, and thus easily detects protein translocation in comparison between control and experimental condition. In our study, medium application of glutamate was found to result in partial PKCδ translocation to mitochondria, a statistically highly significant result based only on a limited number of acquired images.


Subject(s)
Astrocytes/metabolism , Molecular Imaging , Animals , Animals, Newborn , Astrocytes/cytology , Fluorescent Antibody Technique , Mitochondria/metabolism , Molecular Imaging/methods , Protein Kinase C-delta/metabolism , Protein Transport , Rats
5.
J Neuroinflammation ; 14(1): 152, 2017 07 28.
Article in English | MEDLINE | ID: mdl-28754118

ABSTRACT

BACKGROUND: In multiple sclerosis, coagulation factors have been shown to modulate inflammation. In this translational study, we investigated whether long-term anticoagulation with warfarin or rivaroxaban has beneficial effects on the course of autoimmune experimental encephalomyelitis (EAE). METHODS: Female SJL/J mice treated with anticoagulants namely warfarin or rivaroxaban were immunized with PLP139-151. Stable anticoagulation was maintained throughout the entire experiment. Mice without anticoagulation treated with the vehicle only were used as controls. The neurological deficit was recorded during the course of EAE, and histopathological analyses of inflammatory lesions were performed. RESULTS: In preventive settings, both treatment with warfarin and rivaroxaban reduced the maximum EAE score as compared to the control group and led to a reduction of inflammatory lesions in the spinal cord. In contrast, therapeutic treatment with warfarin had no beneficial effects on the clinical course of EAE. Signs of intraparenchymal hemorrhage at the site of the inflammatory lesions were not observed. CONCLUSION: We developed long-term anticoagulation models that allowed exploring the course of EAE under warfarin and rivaroxaban treatment. We found a mild preventive effect of both warfarin and rivaroxaban on neurological deficits and local inflammation, indicating a modulation of the disease induction by anticoagulation.


Subject(s)
Anticoagulants/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Rivaroxaban/therapeutic use , Warfarin/therapeutic use , Animals , Brain/anatomy & histology , Disease Models, Animal , Electric Impedance , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Endothelial Cells/drug effects , Freund's Adjuvant/toxicity , Mice , Myelin Proteolipid Protein/toxicity , Peptide Fragments/toxicity , Random Allocation , Rivaroxaban/blood , Swine , Thrombin/metabolism , Time Factors
6.
J Neurosci Res ; 95(11): 2182-2194, 2017 11.
Article in English | MEDLINE | ID: mdl-28370142

ABSTRACT

Connexin 43 (Cx43) is the main astrocytic connexin and forms the basis of the glial syncytium. The morphology of connexin-expressing cells can be best studied in transgenic mouse lines expressing cytoplasmic fluorescent reporters, since immunolabeling the plaques can obscure the shapes of the individual cells. The Cx43kiECFP mouse generated by Degen et al. (FASEBJ 26:4576, 2012) expresses cytosolic ECFP and has previously been used to establish that Cx43 may not be expressed by all astrocytes within a population, and this can vary in a region-dependent way. To establish this mouse line as a tool for future astrocyte and connexin research, we sought to consolidate reporter authenticity, studying cell types and within-region population heterogeneity. Applying anti-GFP, all cell types related to astroglia were positive-namely, protoplasmic astrocytes in the hippocampus, cortex, thalamus, spinal cord, olfactory bulb, cerebellum with Bergmann glia and astrocytes also in the molecular layer, and retinal Müller cells and astrocytes. Labeled cell types further comprise white matter astrocytes, olfactory ensheathing cells, radial glia-like stem cells, retinal pigment epithelium cells, ependymal cells, and meningeal cells. We furthermore describe a retinal Cx43-expressing amacrine cell morphologically reminiscent of ON-OFF wide-field amacrine cells, representing the first example of a mammalian CNS neuron-expressing Cx43 protein. In double staining with cell type-specific markers (GFAP, S100ß, glutamine synthetase), Cx43 reporter expression in the hippocampus and cortex was restricted to GFAP+ astrocytes. Altogether, this mouse line is a highly reliable tool for studies of Cx43-expressing CNS cells and astroglial cell morphology. © 2017 Wiley Periodicals, Inc.


Subject(s)
Amacrine Cells/metabolism , Connexin 43/biosynthesis , Green Fluorescent Proteins/metabolism , Neuroglia/metabolism , Olfactory Mucosa/cytology , Olfactory Mucosa/metabolism , Amacrine Cells/chemistry , Animals , Connexin 43/analysis , Female , Green Fluorescent Proteins/analysis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroglia/chemistry , Olfactory Bulb/chemistry , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , Olfactory Mucosa/chemistry
7.
Neurochem Res ; 42(6): 1795-1809, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28243788

ABSTRACT

Astrocytes in vivo extend thin processes termed peripheral astrocyte processes (PAPs), in particular around synapses where they can mediate glia-neuronal communication. The relation of PAPs to synapses is not based on coincidence, but it is not clear which stimuli and mechanisms lead to their formation and are active during process extension/ retraction in response to neuronal activity. Also, the molecular basis of the extremely fine PAP morphology (often 50 to 100 nm) is not understood. These open questions can be best investigated under in vitro conditions studying glial filopodia. We have previously analyzed filopodial mechanisms (Lavialle et al. PNAS 108:12915) applying an automated method for filopodia morphometry, which is now described in greater detail. The Filopodia Specific Shape Factor (FSSF) developed integrates number and length of filopodia. It quantifies filopodia independent of overall astrocytic shape or size, which can be intricate in itself. The algorithm supplied here permits automated image processing and measurements using ImageJ. Cells have to be sampled in higher numbers to obtain significant results. We validate the FSSF, and characterize the systematic influence of thresholding and camera pixel grid on measurements. We provide exemplary results of substance-induced filopodia dynamics (glutamate, mGluR agonists, EGF), and show that filopodia formation is highly sensitive to medium pH (CO2) and duration of cell culture. Although the FSSF was developed to study astrocyte filopodia with focus on the perisynaptic glial sheath, we expect that this parameter can also be applied to neuronal growth cones, non-neural cell types, or cell lines.


Subject(s)
Algorithms , Cell Movement/physiology , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Pseudopodia/physiology , Animals , Animals, Newborn , Cell Count/methods , Cells, Cultured , Rats
8.
Glia ; 65(2): 293-308, 2017 02.
Article in English | MEDLINE | ID: mdl-27785828

ABSTRACT

Perivascular endfeet of astrocytes are highly polarized compartments that ensheath blood vessels and contribute to the blood-brain barrier. They experience calcium transients with neuronal activity, a phenomenon involved in neurovascular coupling. Endfeet also mediate the uptake of glucose from the blood, a process stimulated in active brain regions. Here, we demonstrate in mouse hippocampal tissue slices that endfeet undergo sodium signaling upon stimulation of glutamatergic synaptic activity. Glutamate-induced endfeet sodium transients were diminished by TFB-TBOA, suggesting that they were generated by sodium-dependent glutamate uptake. With local agonist application, they could be restricted to endfeet and immunohistochemical analysis revealed prominent expression of glutamate transporters GLAST and GLT-1 localized towards the neuropil vs. the vascular side of endfeet. Endfeet sodium signals spread at an apparent maximum velocity of ∼120 µm/s and directly propagated from stimulated into neighboring endfeet; this spread was omitted in Cx30/Cx43 double-deficient mice. Sodium transients resulted in elevation of intracellular magnesium, indicating a decrease in intracellular ATP. In summary, our results establish that excitatory synaptic activity and stimulation of glutamate uptake in astrocytes trigger transient sodium increases in perivascular endfeet which rapidly spread through gap junctions into neighboring endfeet and cause a reduction of intracellular ATP. The newly discovered endfeet sodium signaling thereby represents a fast, long-lived and inter-cellularly acting indicator of synaptic activity at the blood-brain barrier, which likely constitutes an important component of neuro-metabolic coupling in the brain. GLIA 2017;65:293-308.


Subject(s)
Adenosine Triphosphate/metabolism , Astrocytes/cytology , Gap Junctions/metabolism , Glutamic Acid/metabolism , Signal Transduction/physiology , Sodium/metabolism , Amino Acid Transport System X-AG/antagonists & inhibitors , Animals , Animals, Newborn , Aspartic Acid/analogs & derivatives , Aspartic Acid/pharmacology , Astrocytes/drug effects , Connexin 30/deficiency , Connexin 30/genetics , Connexin 43/deficiency , Connexin 43/genetics , D-Aspartic Acid/pharmacology , Female , Gap Junctions/drug effects , Glutamic Acid/pharmacology , Hippocampus/cytology , Hippocampus/metabolism , Male , Membrane Potentials/drug effects , Mice , Mice, Inbred BALB C , Mice, Knockout , Signal Transduction/drug effects , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
9.
Neuroendocrinology ; 102(4): 311-326, 2015.
Article in English | MEDLINE | ID: mdl-26044072

ABSTRACT

BACKGROUND/AIMS: Cell proliferation and apoptosis are known to adjust neuroendocrine circuits to the photoperiod. The latter is communicated by melatonin, the hormone secreted by the pineal organ. The present study investigated zeitgeber time (ZT)-dependent changes in cell proliferation and apoptosis in the adult murine neuroendocrine system and their regulation by melatonin. METHODS: Adult melatonin-proficient (C3H/HeN) and melatonin-deficient (C57Bl/6J) mice, as well as melatonin-proficient (C3H/HeN) mice with targeted deletion of both melatonin receptor types (MT1 and MT2) were adapted to a 12-hour light, 12-hour dark photoperiod and were sacrificed at ZT00, ZT06, ZT12, and ZT18. Immunohistochemistry for Ki67 and activated caspase-3 served to identify and quantify proliferating and apoptotic cells in the median eminence (ME), hypophyseal pars tuberalis, and pars distalis (PD). RESULTS: ZT-dependent changes in cell proliferation and apoptosis were found exclusively in melatonin-proficient mice with functional MTs. Cell proliferation in the ME and PD showed ZT-dependent changes indicated by an increase at ZT12 (ME) and a decrease at ZT06 (PD). Apoptosis showed ZT-dependent changes in all regions analyzed, indicated by an increase at ZT06. Proliferating and apoptotic cells were found in nearly all cell types residing in the regions analyzed. CONCLUSIONS: Our results indicate that ZT-dependent changes in cell proliferation are counterbalanced by ZT-dependent changes in apoptosis exclusively in melatonin-proficient mice with functional MTs. Melatonin signaling appears to be crucial in both the generation and timing of proliferation and apoptosis that serve the high rate of physiological cell turnover in the adult neuroendocrine system.

10.
Neurochem Res ; 40(12): 2402-13, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25894677

ABSTRACT

The peripheral astrocyte process (PAP) is the glial compartment largely handling inactivation of transmitter glutamate, and supplying glutamate to the axon terminal. It is not clear how these energy demanding processes are fueled, and whether the PAP exhibits oxidative capability. Whereas the GFAP-positive perinuclear cytoplasm and stem process are rich in mitochondria, the PAP is often considered too narrow to contain mitochondria and might thus not rely on oxidative metabolism. Applying high resolution light microscopy, we investigate here the presence of mitochondria in the PAPs of freshly dissociated, isolated astrocytes. We provide an overview of the subcellular distribution and the approximate size of astrocytic mitochondria. A substantial proportion of the astrocyte's mitochondria are contained in the PAPs and, on the average, they are smaller there than in the stem processes. The majority of mitochondria in the stem and peripheral processes are surprisingly small (0.2-0.4 µm), spherical and not elongate, or tubular, which is supported by electron microscopy. The density of mitochondria is two to several times lower in the PAPs than in the stem processes. Thus, PAPs do not constitute a mitochondria free glial compartment but contain mitochondria in large numbers. No juxtaposition of mitochondria-containing PAPs and glutamatergic synapses has been reported. However, the issue of sufficient ATP concentrations in perisynaptic PAPs can be seen in the light of (1) the rapid, activity dependent PAP motility, and (2) the recently reported activity-dependent mitochondrial transport and immobilization leading to spatial, subcellular organisation of glutamate uptake and oxidative metabolism.


Subject(s)
Astrocytes/metabolism , Mitochondria/metabolism , Mitochondrial Size/physiology , Neuroglia/metabolism , Neurotransmitter Agents/metabolism , Adenosine Triphosphate/metabolism , Animals , Astrocytes/ultrastructure , Glial Fibrillary Acidic Protein/metabolism , Mitochondria/ultrastructure , Neural Stem Cells/metabolism , Neuroglia/ultrastructure , Oxidation-Reduction , Peripheral Nervous System/cytology , Peripheral Nervous System/metabolism , Primary Cell Culture , Rats , Subcellular Fractions/metabolism , Synapses/metabolism
12.
PLoS One ; 8(7): e70124, 2013.
Article in English | MEDLINE | ID: mdl-23936150

ABSTRACT

BACKGROUND: The Sphingosine-1-phosphate (S1P) signaling pathway is known to influence pathophysiological processes within the brain and the synthetic S1P analog FTY720 has been shown to provide neuroprotection in experimental models of acute stroke. However, the effects of a manipulation of S1P signaling at later time points after experimental stroke have not yet been investigated. We examined whether a relatively late initiation of a FTY720 treatment has a positive effect on long-term neurological outcome with a focus on reactive astrogliosis, synapses and neurotrophic factors. METHODS: We induced photothrombotic stroke (PT) in adult C57BL/6J mice and allowed them to recover for three days. Starting on post-stroke day 3, mice were treated with FTY720 (1 mg/kg b.i.d.) for 5 days. Behavioral outcome was observed until day 31 after photothrombosis and periinfarct cortical tissue was analyzed using tandem mass-spectrometry, TaqMan®analysis and immunofluorescence. RESULTS: FTY720 treatment results in a significantly better functional outcome persisting up to day 31 after PT. This is accompanied by a significant decrease in reactive astrogliosis and larger post-synaptic densities as well as changes in the expression of vascular endothelial growth factor α (VEGF α). Within the periinfarct cortex, S1P is significantly increased compared to healthy brain tissue. CONCLUSION: Besides its known neuroprotective effects in the acute phase of experimental stroke, the initiation of FTY720 treatment in the convalescence period has a positive impact on long-term functional outcome, probably mediated through reduced astrogliosis, a modulation in synaptic morphology and an increased expression of neurotrophic factors.


Subject(s)
Gliosis/prevention & control , Propylene Glycols/pharmacology , Recovery of Function/drug effects , Sphingosine/analogs & derivatives , Stroke/prevention & control , Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism , Animals , Astrocytes/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Convalescence , Fingolimod Hydrochloride , Gene Expression/drug effects , Gliosis/physiopathology , Immunosuppressive Agents/metabolism , Immunosuppressive Agents/pharmacology , Intracranial Thrombosis/complications , Male , Mice , Mice, Inbred C57BL , Post-Synaptic Density/drug effects , Propylene Glycols/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sphingosine/metabolism , Sphingosine/pharmacology , Stroke/etiology , Stroke/physiopathology , Time Factors , Treatment Outcome , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
13.
Front Cell Neurosci ; 7: 54, 2013.
Article in English | MEDLINE | ID: mdl-23653590

ABSTRACT

Studying the distribution of astrocytic antigens is particularly hard when they are localized in their fine, peripheral astrocyte processes (PAPs), since these processes often have a diameter comparable to vesicles and small organelles. The most appropriate technique is immunoelectron microscopy, which is, however, a time-consuming procedure. Even in high resolution light microscopy, antigen localization is difficult to detect due to the small dimensions of these processes, and overlay from antigen in surrounding non-glial cells. Yet, PAPs frequently display antigens related to motility and glia-synaptic interaction. Here, we describe the dissociation of morphologically intact glial cells (DIMIGs), permitting unambiguous antigen localization using epifluorescence microscopy. Astrocytes are dissociated from juvenile (p13-15) mouse cortex by applying papain treatment and cytospin centrifugation to attach the cells to a slide. The cells and their complete processes including the PAPs is thus projected in 2D. The entire procedure takes 2.5-3 h. We show by morphometry that the diameter of DIMIGs, including the PAPs is similar to that of astrocytes in situ. In contrast to cell culture, results derived from this procedure allow for direct conclusions relating to (1) the presence of an antigen in cortical astrocytes, (2) subcellular antigen distribution, in particular when localized in the PAPs. The detailed resolution is shown in an exemplary study of the organization of the astrocytic cytoskeleton components actin, ezrin, tubulin, and GFAP. The distribution of connexin 43 in relation to a single astrocyte's process tree is also investigated.

14.
Neurochem Res ; 37(11): 2513-23, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22730011

ABSTRACT

Various ependymoglial cells display varying degrees of process specialization, in particular processes contacting mesenchymal borders (pia, blood vessels, vitreous body), or those lining the ventricular surface. Within the neuropil, glial morphology, cellular contacts, and interaction partners are complex. It appears that glial processes contacting neurons, specific parts of neurons, or mesenchymal or ventricular borders are characterized by specialized membranes. We propose a concept of membrane domains in addition to the existing concept of ependymoglial polarity. Such membrane domains are equipped with certain membrane-bound proteins, enabling them to function in their specific environment. This review focuses on Müller cells and astrocytes and discusses exemplary the localization of established glial markers in membrane domains. We distinguish three functional glial membrane domains based on their typical molecular arrangement. The domain of the endfoot specifically displays the complex of dystrophin-associated proteins, aquaporin 4 and the potassium channel Kir4.1. We show that the domain of microvilli and the peripheral glial process in the Müller cell share the presence of ezrin, as do peripheral astrocyte processes. As a third domain, the Müller cell has peripheral glial processes related to a specific subtype of synapse. Although many details remain to be studied, the idea of glial membrane domains may permit new insights into glial function and pathology.


Subject(s)
Astrocytes/cytology , Cell Polarity , Neuroglia/metabolism , Animals , Humans , Nerve Tissue Proteins/metabolism
15.
Cell Mol Life Sci ; 69(3): 461-70, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21779868

ABSTRACT

While the availability of pluripotent stem cells has opened new prospects for generating neural donor cells for nervous system repair, their capability to integrate with adult brain tissue in a structurally relevant way is still largely unresolved. We addressed the potential of human embryonic stem cell-derived long-term self-renewing neuroepithelial stem cells (lt-NES cells) to establish axonal projections after transplantation into the adult rodent brain. Transgenic and species-specific markers were used to trace the innervation pattern established by transplants in the hippocampus and motor cortex. In vitro, lt-NES cells formed a complex axonal network within several weeks after the initiation of differentiation and expressed a composition of surface receptors known to be instrumental in axonal growth and pathfinding. In vivo, these donor cells adopted projection patterns closely mimicking endogenous projections in two different regions of the adult rodent brain. Hippocampal grafts placed in the dentate gyrus projected to both the ipsilateral and contralateral pyramidal cell layers, while axons of donor neurons placed in the motor cortex extended via the external and internal capsule into the cervical spinal cord and via the corpus callosum into the contralateral cortex. Interestingly, acquisition of these region-specific projection profiles was not correlated with the adoption of a regional phenotype. Upon reaching their destination, human axons established ultrastructural correlates of synaptic connections with host neurons. Together, these data indicate that neurons derived from human pluripotent stem cells are endowed with a remarkable potential to establish orthotopic long-range projections in the adult mammalian brain.


Subject(s)
Embryonic Stem Cells/cytology , Neurons/cytology , Animals , Axons/physiology , Brain Tissue Transplantation , Cells, Cultured , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/transplantation , Humans , Neurogenesis , Neurons/transplantation , Rats , Rats, Sprague-Dawley , Synapses/pathology , Synapses/ultrastructure
16.
Proc Natl Acad Sci U S A ; 108(31): 12915-9, 2011 Aug 02.
Article in English | MEDLINE | ID: mdl-21753079

ABSTRACT

The peripheral astrocyte process (PAP) preferentially associates with the synapse. The PAP, which is not found around every synapse, extends to or withdraws from it in an activity-dependent manner. Although the pre- and postsynaptic elements have been described in great molecular detail, relatively little is known about the PAP because of its difficult access for electrophysiology or light microscopy, as they are smaller than microscopic resolution. We investigated possible stimuli and mechanisms of PAP plasticity. Immunocytochemistry on rat brain sections demonstrates that the actin-binding protein ezrin and the metabotropic glutamate receptors (mGluRs) 3 and 5 are compartmentalized to the PAP but not to the GFAP-containing stem process. Further experiments applying ezrin siRNA or dominant-negative ezrin in primary astrocytes indicate that filopodia formation and motility require ezrin in the membrane/cytoskeleton bound (i.e., T567-phosphorylated) form. Glial processes around synapses in situ consistently display this ezrin form. Possible motility stimuli of perisynaptic glial processes were studied in culture, based on their similarity with filopodia. Glutamate and glutamate analogues reveal that rapid (5 min), glutamate-induced filopodia motility is mediated by mGluRs 3 and 5. Ultrastructurally, these mGluR subtypes were also localized in astrocytes in the rat hippocampus, preferentially in their fine PAPs. In vivo, changes in glutamatergic circadian activity in the hamster suprachiasmatic nucleus are accompanied by changes of ezrin immunoreactivity in the suprachiasmatic nucleus, in line with transmitter-induced perisynaptic glial motility. The data suggest that (i) ezrin is required for the structural plasticity of PAPs and (ii) mGluRs can stimulate PAP plasticity.


Subject(s)
Astrocytes/metabolism , Cytoskeletal Proteins/metabolism , Receptors, Metabotropic Glutamate/metabolism , Synapses/physiology , Animals , Astrocytes/cytology , Astrocytes/ultrastructure , Cells, Cultured , Cricetinae , Cytoskeletal Proteins/genetics , Female , Glial Fibrillary Acidic Protein/metabolism , Glutamic Acid/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Immunohistochemistry , Male , Mesocricetus , Microscopy, Fluorescence , Microscopy, Immunoelectron , Neuronal Plasticity/physiology , Pregnancy , Pseudopodia/drug effects , Pseudopodia/metabolism , Pseudopodia/physiology , RNA Interference , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5 , Synapses/metabolism
17.
Neural Dev ; 6: 29, 2011 Jun 20.
Article in English | MEDLINE | ID: mdl-21689430

ABSTRACT

BACKGROUND: The ventral midbrain contains a diverse array of neurons, including dopaminergic neurons of the ventral tegmental area (VTA) and substantia nigra (SN) and neurons of the red nucleus (RN). Dopaminergic and RN neurons have been shown to arise from ventral mesencephalic precursors that express Sonic Hedgehog (Shh). However, Shh expression, which is initially confined to the mesencephalic ventral midline, expands laterally and is then downregulated in the ventral midline. In contrast, expression of the Hedgehog target gene Gli1 initiates in the ventral midline prior to Shh expression, but after the onset of Shh expression it is expressed in precursors lateral to Shh-positive cells. Given these dynamic gene expression patterns, Shh and Gli1 expression could delineate different progenitor populations at distinct embryonic time points. RESULTS: We employed genetic inducible fate mapping (GIFM) to investigate whether precursors that express Shh (Shh-GIFM) or transduce Shh signaling (Gli1-GIFM) at different time points give rise to different ventral midbrain cell types. We find that precursors restricted to the ventral midline are labeled at embryonic day (E)7.5 with Gli1-GIFM, and with Shh-GIFM at E8.5. These precursors give rise to all subtypes of midbrain dopaminergic neurons and the anterior RN. A broader domain of progenitors that includes the ventral midline is marked with Gli1-GIFM at E8.5 and with Shh-GIFM at E9.5; these fate-mapped cells also contribute to all midbrain dopaminergic subtypes and to the entire RN. In contrast, a lateral progenitor domain that is labeled with Gli1-GIFM at E9.5 and with Shh-GIFM at E11.5 has a markedly reduced potential to give rise to the RN and to SN dopaminergic neurons, and preferentially gives rise to the ventral-medial VTA. In addition, cells derived from Shh- and Gli1-expressing progenitors located outside of the ventral midline give rise to astrocytes. CONCLUSIONS: We define a ventral midbrain precursor map based on the timing of Gli1 and Shh expression, and suggest that the diversity of midbrain dopaminergic neurons is at least partially determined during their precursor stage when their medial-lateral position, differential gene expression and the time when they leave the ventricular zone influence their fate decisions.


Subject(s)
Hedgehog Proteins/genetics , Hedgehog Proteins/physiology , Mesencephalon/physiology , Neural Stem Cells/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Animals , Astrocytes/physiology , Brain Mapping , Cell Differentiation/genetics , Dopamine/physiology , Female , Fluorescent Antibody Technique , In Situ Hybridization , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/physiology , Mesencephalon/cytology , Mesencephalon/embryology , Mice , Neurons/physiology , Oculomotor Nerve/embryology , Oculomotor Nerve/growth & development , Pregnancy , RNA/biosynthesis , RNA/genetics , Red Nucleus/cytology , Red Nucleus/embryology , Red Nucleus/physiology , Substantia Nigra/embryology , Substantia Nigra/growth & development , Substantia Nigra/physiology , Zinc Finger Protein GLI1
18.
PLoS One ; 6(3): e17575, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21455301

ABSTRACT

NG2 cells, the fourth type of glia in the mammalian CNS, receive synaptic input from neurons. The function of this innervation is unknown yet. Postsynaptic changes in intracellular Ca(2+)-concentration ([Ca(2+)](i)) might be a possible consequence. We employed transgenic mice with fluorescently labeled NG2 cells to address this issue. To identify Ca(2+)-signaling pathways we combined patch-clamp recordings, Ca(2+)-imaging, mRNA-transcript analysis and focal pressure-application of various substances to identified NG2-cells in acute hippocampal slices. We show that activation of voltage-gated Ca(2+)-channels, Ca(2+)-permeable AMPA-receptors, and group I metabotropic glutamate-receptors provoke [Ca(2+)](i)-elevations in NG2 cells. The Ca(2+)-influx is amplified by Ca(2+)-induced Ca(2+)-release. Minimal electrical stimulation of presynaptic neurons caused postsynaptic currents but no somatic [Ca(2+)](i) elevations, suggesting that [Ca(2+)](i) elevations in NG2 cells might be restricted to their processes. Local Ca(2+)-signaling might provoke transmitter release or changes in cell motility. To identify structural prerequisites for such a scenario, we used electron microscopy, immunostaining, mRNA-transcript analysis, and time lapse imaging. We found that NG2 cells form symmetric and asymmetric synapses with presynaptic neurons and show immunoreactivity for vesicular glutamate transporter 1. The processes are actin-based, contain ezrin but not glial filaments, microtubules or endoplasmic reticulum. Furthermore, we demonstrate that NG2 cell processes in situ are highly motile. Our findings demonstrate that gray matter NG2 cells are endowed with the cellular machinery for two-way communication with neighboring cells.


Subject(s)
Calcium/metabolism , Neuroglia/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Electrophysiology , Humans , Mice , Mice, Transgenic , Microscopy, Electron , Microscopy, Fluorescence , Neuroglia/ultrastructure , Reverse Transcriptase Polymerase Chain Reaction , Vesicular Glutamate Transport Protein 1/metabolism
19.
Glia ; 58(13): 1553-69, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20549747

ABSTRACT

The dentate gyrus is a brain region where neurons are continuously born throughout life. In the adult, the role of its radial glia in neurogenesis has attracted much attention over the past years; however, little is known about the generation and differentiation of glial cells and their relationship to radial glia during the ontogenetic development of this brain structure. Here, we combine immunohistochemical phenotyping using antibodies against glial marker proteins with BrdU birthdating to characterize the development of the secondary radial glial scaffold in the dentate gyrus and its potential to differentiate into astrocytes. We demonstrate that the expression of brain lipid-binding protein, GLAST, and glial fibrillary acidic protein (GFAP) characterizes immature differentiating cells confined to an astrocytic fate in the early postnatal dentate gyrus. On the basis of our studies, we propose a model where immature astrocytes migrate radially through the granule cell layer to adopt their final positions in the molecular layer of the dentate gyrus. Time-lapse imaging of acute hippocampal slices from hGFAP-eGFP transgenic mice provides direct evidence for such a migration mode of differentiating astroglial cells in the developing dentate gyrus.


Subject(s)
Dentate Gyrus , Gene Expression Regulation, Developmental/physiology , Neuroglia/physiology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Cell Differentiation/physiology , Cell Proliferation , Dentate Gyrus/cytology , Dentate Gyrus/embryology , Dentate Gyrus/growth & development , Embryo, Mammalian , Excitatory Amino Acid Transporter 1/metabolism , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , In Vitro Techniques , Mice , Mice, Transgenic , Models, Biological , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Time Factors
20.
Brain Res Rev ; 63(1-2): 11-25, 2010 May.
Article in English | MEDLINE | ID: mdl-20176054

ABSTRACT

The major glial population of the brain is constituted by astroglia. Highly branched and ramified protoplasmic astrocytes are the predominant form in grey matter and are found in almost all regions of the central nervous system. In cerebellum and retina, there two forms of elongated radial glia exist (Bergmann glia and Müller cells, respectively) that share many features with the protoplasmic astrocytes in respect to their perisynaptic association. Although these three astroglial cell types are different in their gross morphology, they are characterized by a polarized orientation of their processes. While one or only few processes have contacts with CNS boundaries such as capillaries and pia, an overwhelming number of thin filopodia- and lamellipodia-like process terminals contact and enwrap synapses, the sites of neuronal communication. The perisynaptic glial processes are the primary compartments that sense neuronal activity. After signal integration, they can also modulate synaptic transmission, thereby contributing to neural plasticity. Despite their importance, the mechanisms that (1) target astroglial processes toward pre- and postsynaptic compartments and (2) control the interaction during plastic events of the brain such as learning or injury are poorly understood. This review will summarize our current knowledge and highlight some open questions.


Subject(s)
Neuroglia/cytology , Neuroglia/physiology , Synapses/physiology , Animals , Humans , Neuroglia/ultrastructure , Video Recording
SELECTION OF CITATIONS
SEARCH DETAIL
...