Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Neurochem ; 165(5): 701-721, 2023 06.
Article in English | MEDLINE | ID: mdl-36636908

ABSTRACT

Neuron-restrictive silencer factor/repressor element 1 (RE1)-silencing transcription factor (NRSF/REST) is a transcriptional repressor of a large cluster of neural genes containing RE1 motifs in their promoter region. NRSF/REST is ubiquitously expressed in non-neuronal cells, including astrocytes, while it is down-regulated during neuronal differentiation. While neuronal NRSF/REST homeostatically regulates intrinsic excitability and synaptic transmission, the role of the high NRSF/REST expression levels in the homeostatic functions of astrocytes is poorly understood. Here, we investigated the functional consequences of NRSF/REST deletion in primary cortical astrocytes derived from NRSF/REST conditional knockout mice (KO). We found that NRSF/REST KO astrocyte displayed a markedly reduced activity of inward rectifying K+ channels subtype 4.1 (Kir4.1) underlying spatial K+ buffering that was associated with a decreased expression and activity of the glutamate transporter-1 (GLT-1) responsible for glutamate uptake by astrocytes. The effects of the impaired astrocyte homeostatic functions on neuronal activity were investigated by co-culturing wild-type hippocampal neurons with NRSF/REST KO astrocytes. Interestingly, neurons experienced increased neuronal excitability at high firing rates associated with decrease after hyperpolarization and increased amplitude of excitatory postsynaptic currents. The data indicate that astrocytic NRSF/REST directly participates in neural circuit homeostasis by regulating intrinsic excitability and excitatory transmission and that dysfunctions of NRSF/REST expression in astrocytes may contribute to the pathogenesis of neurological disorders.


Subject(s)
Astrocytes , Transcription Factors , Mice , Animals , Transcription Factors/genetics , Astrocytes/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Gene Expression Regulation
2.
Pflugers Arch ; 474(2): 243-260, 2022 02.
Article in English | MEDLINE | ID: mdl-34734327

ABSTRACT

The capacity of astrocytes to adapt their biochemical and functional features upon physiological and pathological stimuli is a fundamental property at the basis of their ability to regulate the homeostasis of the central nervous system (CNS). It is well known that in primary cultured astrocytes, the expression of plasma membrane ion channels and transporters involved in homeostatic tasks does not closely reflect the pattern observed in vivo. The individuation of culture conditions that promote the expression of the ion channel array found in vivo is crucial when aiming at investigating the mechanisms underlying their dynamics upon various physiological and pathological stimuli. A chemically defined medium containing growth factors and hormones (G5) was previously shown to induce the growth, differentiation, and maturation of primary cultured astrocytes. Here we report that under these culture conditions, rat cortical astrocytes undergo robust morphological changes acquiring a multi-branched phenotype, which develops gradually during the 2-week period of culturing. The shape changes were paralleled by variations in passive membrane properties and background conductance owing to the differential temporal development of inwardly rectifying chloride (Cl-) and potassium (K+) currents. Confocal and immunoblot analyses showed that morphologically differentiated astrocytes displayed a large increase in the expression of the inward rectifier Cl- and K+ channels ClC-2 and Kir4.1, respectively, which are relevant ion channels in vivo. Finally, they exhibited a large diminution of the intermediate filaments glial fibrillary acidic protein (GFAP) and vimentin which are upregulated in reactive astrocytes in vivo. Taken together the data indicate that long-term culturing of cortical astrocytes in this chemical-defined medium promotes a quiescent functional phenotype. This culture model could aid to address the regulation of ion channel expression involved in CNS homeostasis in response to physiological and pathological challenges.


Subject(s)
Astrocytes/metabolism , Homeostasis/physiology , Animals , CLC-2 Chloride Channels/metabolism , Cell Membrane/metabolism , Central Nervous System/metabolism , Central Nervous System/physiology , Chlorides/metabolism , Patch-Clamp Techniques/methods , Potassium/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Rats , Rats, Sprague-Dawley , Vimentin/metabolism
4.
J Cell Sci ; 134(16)2021 08 15.
Article in English | MEDLINE | ID: mdl-34279618

ABSTRACT

Astroglial cells are key to maintain nervous system homeostasis. Neurotrophins are known for their pleiotropic effects on neuronal physiology but also exert complex functions to glial cells. Here, we investigated (i) the signaling competence of mouse embryonic and postnatal primary cortical astrocytes exposed to brain-derived neurotrophic factor (BDNF) and, (ii) the role of kinase D-interacting substrate of 220 kDa (Kidins220), a transmembrane scaffold protein that mediates neurotrophin signaling in neurons. We found a shift from a kinase-based response in embryonic cells to a response predominantly relying on intracellular Ca2+ transients [Ca2+]i within postnatal cultures, associated with a decrease in the synthesis of full-length BDNF receptor TrkB, with Kidins220 contributing to the BDNF-activated kinase and [Ca2+]i pathways. Finally, Kidins220 participates in the homeostatic function of astrocytes by controlling the expression of the ATP-sensitive inward rectifier potassium channel 10 (Kir4.1) and the metabolic balance of embryonic astrocytes. Overall, our data contribute to the understanding of the complex role played by astrocytes within the central nervous system, and identify Kidins220 as a novel actor in the increasing number of pathologies characterized by astrocytic dysfunctions. This article has an associated First Person interview with the first authors of the paper.


Subject(s)
Astrocytes , Brain-Derived Neurotrophic Factor/metabolism , Membrane Proteins/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Cells, Cultured , Mice , Neurons , Signal Transduction
5.
Cell Death Differ ; 27(5): 1505-1519, 2020 05.
Article in English | MEDLINE | ID: mdl-31624352

ABSTRACT

Through their ability to modulate synaptic transmission, glial cells are key regulators of neuronal circuit formation and activity. Kidins220/ARMS (kinase-D interacting substrate of 220 kDa/ankyrin repeat-rich membrane spanning) is one of the key effectors of the neurotrophin pathways in neurons where it is required for differentiation, survival, and plasticity. However, its role in glial cells remains largely unknown. Here, we show that ablation of Kidins220 in primary cultured astrocytes induced defects in calcium (Ca2+) signaling that were linked to altered store-operated Ca2+ entry and strong overexpression of the transient receptor potential channel TRPV4. Moreover, Kidins220-/- astrocytes were more sensitive to genotoxic stress. We also show that Kidins220 expression in astrocytes is required for the establishment of proper connectivity of cocultured wild-type neurons. Altogether, our data reveal a previously unidentified role for astrocyte-expressed Kidins220 in the control of glial Ca2+ dynamics, survival/death pathways and astrocyte-neuron communication.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , Calcium Signaling , Cell Communication , Membrane Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Animals , Calcium/metabolism , Cells, Cultured , DNA Damage , Embryo, Mammalian/cytology , Humans , Mice, Inbred C57BL , Models, Biological , Neurogenesis , TRPV Cation Channels/metabolism
6.
Small ; 15(15): e1900147, 2019 04.
Article in English | MEDLINE | ID: mdl-30891923

ABSTRACT

The use of graphene nanomaterials (GNMs) for biomedical applications targeted to the central nervous system is exponentially increasing, although precise information on their effects on brain cells is lacking. In this work, the molecular changes induced in cortical astrocytes by few-layer graphene (FLG) and graphene oxide (GO) flakes are addressed. The results show that exposure to FLG/GO does not affect cell viability or proliferation. However, proteomic and lipidomic analyses unveil alterations in several cellular processes, including intracellular Ca2+ ([Ca2+ ]i ) homeostasis and cholesterol metabolism, which are particularly intense in cells exposed to GO. Indeed, GO exposure impairs spontaneous and evoked astrocyte [Ca2+ ]i signals and induces a marked increase in membrane cholesterol levels. Importantly, cholesterol depletion fully rescues [Ca2+ ]i dynamics in GO-treated cells, indicating a causal relationship between these GO-mediated effects. The results indicate that exposure to GNMs alters intracellular signaling in astrocytes and may impact astrocyte-neuron interactions.


Subject(s)
Astrocytes/metabolism , Calcium/metabolism , Cell Membrane/metabolism , Cholesterol/metabolism , Graphite/pharmacology , Homeostasis , Animals , Astrocytes/cytology , Astrocytes/drug effects , Calcium Signaling/drug effects , Cell Cycle/drug effects , Cell Membrane/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Homeostasis/drug effects , Intracellular Space/metabolism , Lipidomics , Proteome/metabolism , Rats, Sprague-Dawley
7.
Nano Lett ; 18(9): 5827-5838, 2018 09 12.
Article in English | MEDLINE | ID: mdl-30088941

ABSTRACT

Graphene-based materials are the focus of intense research efforts to devise novel theranostic strategies for targeting the central nervous system. In this work, we have investigated the consequences of long-term exposure of primary rat astrocytes to pristine graphene (GR) and graphene oxide (GO) flakes. We demonstrate that GR/GO interfere with a variety of intracellular processes as a result of their internalization through the endolysosomal pathway. Graphene-exposed astrocytes acquire a more differentiated morphological phenotype associated with extensive cytoskeletal rearrangements. Profound functional alterations are induced by GO internalization, including the upregulation of inward-rectifying K+ channels and of Na+-dependent glutamate uptake, which are linked to the astrocyte capacity to control the extracellular homeostasis. Interestingly, GO-pretreated astrocytes promote the functional maturation of cocultured primary neurons by inducing an increase in intrinsic excitability and in the density of GABAergic synapses. The results indicate that graphene nanomaterials profoundly affect astrocyte physiology in vitro with consequences for neuronal network activity. This work supports the view that GO-based materials could be of great interest to address pathologies of the central nervous system associated with astrocyte dysfunctions.


Subject(s)
Astrocytes/cytology , Graphite/metabolism , Neurons/cytology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cell Communication/drug effects , Cell Shape/drug effects , Cells, Cultured , Glutamic Acid/metabolism , Graphite/chemistry , Homeostasis/drug effects , Nanostructures/chemistry , Neurons/drug effects , Neurons/metabolism , Potassium Channels/metabolism , Rats , Synapses/metabolism
8.
Stem Cell Reports ; 4(1): 25-36, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25556566

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.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , Cell Transdifferentiation/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Transcription Factors/genetics , Animals , Astrocytes/drug effects , Biomarkers , Cell Transdifferentiation/drug effects , Cells, Cultured , Cellular Reprogramming/genetics , Cluster Analysis , Cytokines/metabolism , Cytokines/pharmacology , Fibroblasts/drug effects , Gene Expression , Gene Expression Profiling , Humans , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Phenotype , Transcription Factors/metabolism
9.
J Neurochem ; 132(3): 286-300, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25279950

ABSTRACT

Accumulating evidence indicates that increased intracellular Na(+) concentration ([Na(+) ]i ) in astroglial cells is associated with the development of brain edema under ischemic conditions, but the underlying mechanisms are still elusive. Here, we report that in primary cultured rat cortical astrocytes, elevations of [Na(+) ]i reflecting those achieved during ischemia cause a marked decrease in hypotonicity-evoked current mediated by volume-regulated anion channel (VRAC). Pharmacological manipulations revealed that VRAC inhibition was not due to the reverse mode of the plasma membrane sodium/calcium exchanger. The negative modulation of VRAC was also observed in an astrocytic cell line lacking the predominant astrocyte water channel aquaporin 4, indicating that [Na(+) ]i effect was not mediated by the regulation of aquaporin 4 activity. The inward rectifier Cl(-) current, which is also expressed by cultured astrocytes, was not affected by [Na(+) ]i increase. VRAC depression by high [Na(+) ]i was confirmed in adult astrocytes, suggesting that it was not developmentally regulated. Altogether, these results provide the first evidence that intracellular Na(+) dynamics can modulate astrocytic membrane conductance that controls functional processes linked to cell volume regulation and add further support to the concept that limiting astrocyte intracellular Na(+) accumulation might be a favorable strategy to counteract the development of brain edema.


Subject(s)
Astrocytes/metabolism , Cerebral Cortex/metabolism , Ion Channels/drug effects , Sodium/pharmacology , Animals , Aquaporin 4/metabolism , Astrocytes/drug effects , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Chloride Channels/metabolism , Female , Male , Patch-Clamp Techniques , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Sodium/metabolism , Sodium-Calcium Exchanger/metabolism
10.
Hum Mol Genet ; 23(19): 5069-86, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24824219

ABSTRACT

Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a leukodystrophy characterized by myelin vacuolization and caused by mutations in MLC1 or GLIALCAM. Patients with recessive mutations in either MLC1 or GLIALCAM show the same clinical phenotype. It has been shown that GLIALCAM is necessary for the correct targeting of MLC1 to the membrane at cell junctions, but its own localization was independent of MLC1 in vitro. However, recent studies in Mlc1(-/-) mice have shown that GlialCAM is mislocalized in glial cells. In order to investigate whether the relationship between Mlc1 and GlialCAM is species-specific, we first identified MLC-related genes in zebrafish and generated an mlc1(-/-) zebrafish. We have characterized mlc1(-/-) zebrafish both functionally and histologically and compared the phenotype with that of the Mlc1(-/-) mice. In mlc1(-/-) zebrafish, as in Mlc1(-/-) mice, Glialcam is mislocalized. Re-examination of a brain biopsy from an MLC patient indicates that GLIALCAM is also mislocalized in Bergmann glia in the cerebellum. In vitro, impaired localization of GlialCAM was observed in astrocyte cultures from Mlc1(-/-) mouse only in the presence of elevated potassium levels, which mimics neuronal activity. In summary, here we demonstrate an evolutionary conserved role for MLC1 in regulating glial surface levels of GLIALCAM, and this interrelationship explains why patients with mutations in either gene (MLC1 or GLIALCAM) share the same clinical phenotype.


Subject(s)
Cysts/metabolism , Hereditary Central Nervous System Demyelinating Diseases/metabolism , Membrane Proteins/metabolism , Neuroglia/metabolism , Proteins/metabolism , Animals , Animals, Genetically Modified , Astrocytes/metabolism , Brain/metabolism , Brain/pathology , Cell Cycle Proteins , Cell Line , Cell Membrane/metabolism , Cysts/genetics , Disease Models, Animal , Ependyma/cytology , Ependyma/metabolism , Ependyma/ultrastructure , Gene Expression , Genotype , Hereditary Central Nervous System Demyelinating Diseases/genetics , Humans , Intercellular Junctions/metabolism , Intercellular Junctions/ultrastructure , Membrane Proteins/genetics , Mice , Mice, Knockout , Mutation , Phenotype , Protein Transport , Proteins/genetics , Retina/metabolism , Voltage-Dependent Anion Channels/metabolism , Zebrafish/genetics , Zebrafish/metabolism
11.
Adv Healthc Mater ; 3(3): 392-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23966220

ABSTRACT

Astroglial ion channels are fundamental molecular targets in the study of brain physiology and pathophysiology. Novel tools and devices intended for stimulation and control of astrocytes ion channel activity are therefore highly desirable. The study of the interactions between astrocytes and biomaterials is also essential to control and minimize reactive astrogliosis, in view of the development of implantable functional devices. Here, the growth of rat primary neocortical astrocytes on the top of a light sensitive, organic polymer film is reported; by means of patch-clamp analyses, the effect of the visible light stimulation on membrane conductance is then determined. Photoexcitation of the active material causes a significant depolarization of the astroglial resting membrane potential: the effect is associated to an increase in whole-cell conductance at negative potentials. The magnitude of the evoked inward current density is proportional to the illumination intensity. Biophysical and pharmacological characterization suggests that the ion channel mediating the photo-transduction mechanism is a chloride channel, the ClC-2 channel. These results open interesting perspectives for the selective manipulation of astrocyte bioelectrical activity by non-invasive, label-free, organic-based, photostimulation approaches.


Subject(s)
Astrocytes/radiation effects , Light , Neocortex/chemistry , Polymers/chemistry , Animals , CLC-2 Chloride Channels , Cell Survival/radiation effects , Cells, Cultured , Chloride Channels/metabolism , Membrane Potentials/radiation effects , Rats
12.
PLoS One ; 7(6): e39959, 2012.
Article in English | MEDLINE | ID: mdl-22761937

ABSTRACT

The polymodal transient receptor potential vanilloid 4 (TRPV4) channel, a member of the TRP channel family, is a calcium-permeable cationic channel that is gated by various stimuli such as cell swelling, low pH and high temperature. Therefore, TRPV4-mediated calcium entry may be involved in neuronal and glia pathophysiology associated with various disorders of the central nervous system, such as ischemia. The TRPV4 channel has been recently found in adult rat cortical and hippocampal astrocytes; however, its role in astrocyte pathophysiology is still not defined. In the present study, we examined the impact of cerebral hypoxia/ischemia (H/I) on the functional expression of astrocytic TRPV4 channels in the adult rat hippocampal CA1 region employing immunohistochemical analyses, the patch-clamp technique and microfluorimetric intracellular calcium imaging on astrocytes in slices as well as on those isolated from sham-operated or ischemic hippocampi. Hypoxia/ischemia was induced by a bilateral 15-minute occlusion of the common carotids combined with hypoxic conditions. Our immunohistochemical analyses revealed that 7 days after H/I, the expression of TRPV4 is markedly enhanced in hippocampal astrocytes of the CA1 region and that the increasing TRPV4 expression coincides with the development of astrogliosis. Additionally, adult hippocampal astrocytes in slices or cultured hippocampal astrocytes respond to the TRPV4 activator 4-alpha-phorbol-12,-13-didecanoate (4αPDD) by an increase in intracellular calcium and the activation of a cationic current, both of which are abolished by the removal of extracellular calcium or exposure to TRP antagonists, such as Ruthenium Red or RN1734. Following hypoxic/ischemic injury, the responses of astrocytes to 4αPDD are significantly augmented. Collectively, we show that TRPV4 channels are involved in ischemia-induced calcium entry in reactive astrocytes and thus, might participate in the pathogenic mechanisms of astroglial reactivity following ischemic insult.


Subject(s)
Astrocytes/physiology , Hippocampus/physiopathology , Hypoxia-Ischemia, Brain/physiopathology , TRPV Cation Channels/physiology , Animals , Base Sequence , Blotting, Western , DNA Primers , Hippocampus/pathology , Hypoxia-Ischemia, Brain/pathology , Immunohistochemistry , Male , Patch-Clamp Techniques , Polymerase Chain Reaction , Rats , Rats, Wistar
14.
Proc Natl Acad Sci U S A ; 108(6): 2563-8, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21262839

ABSTRACT

Regulatory volume decrease (RVD) is a key mechanism for volume control that serves to prevent detrimental swelling in response to hypo-osmotic stress. The molecular basis of RVD is not understood. Here we show that a complex containing aquaporin-4 (AQP4) and transient receptor potential vanilloid 4 (TRPV4) is essential for RVD in astrocytes. Astrocytes from AQP4-KO mice and astrocytes treated with TRPV4 siRNA fail to respond to hypotonic stress by increased intracellular Ca(2+) and RVD. Coimmunoprecipitation and immunohistochemistry analyses show that AQP4 and TRPV4 interact and colocalize. Functional analysis of an astrocyte-derived cell line expressing TRPV4 but not AQP4 shows that RVD and intracellular Ca(2+) response can be reconstituted by transfection with AQP4 but not with aquaporin-1. Our data indicate that astrocytes contain a TRPV4/AQP4 complex that constitutes a key element in the brain's volume homeostasis by acting as an osmosensor that couples osmotic stress to downstream signaling cascades.


Subject(s)
Aquaporin 4/metabolism , Astrocytes/metabolism , Cell Size , TRPV Cation Channels/metabolism , Animals , Aquaporin 1/genetics , Aquaporin 1/metabolism , Aquaporin 4/genetics , Astrocytes/cytology , COS Cells , Calcium/metabolism , Chlorocebus aethiops , Cricetinae , Humans , Mice , Mice, Knockout , Osmotic Pressure/physiology , Signal Transduction/physiology , TRPV Cation Channels/genetics
15.
Biomaterials ; 31(31): 7883-91, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20688390

ABSTRACT

Astroglial cell survival and ion channel activity are relevant molecular targets for the mechanistic study of neural cell interactions with biomaterials and/or electronic interfaces. Astrogliosis is the most typical reaction to in vivo brain implants and needs to be avoided by developing biomaterials that preserve astroglial cell physiological function. This cellular phenomenon is characterized by a proliferative state and altered expression of astroglial potassium (K(+)) channels. Silk is a natural polymer with potential for new biomedical applications due to its ability to support in vitro growth and differentiation of many cell types. We report on silk interactions with cultured neocortical astroglial cells. Astrocytes survival is similar when plated on silk-coated glass and on poly-D-lysine (PDL), a well known polyionic substrate used to promote astroglial cell adhesion to glass surfaces. Comparative analyses of whole-cell patch-clamp experiments reveal that silk- and PDL-coated cells display depolarized resting membrane potentials (-40 mV), very high input resistance, and low specific conductance, with values similar to those of undifferentiated glial cells. Analysis of K(+) channel conductance reveals that silk-astrocytes express large outwardly delayed rectifying K(+) current (K(DR)). The magnitude of K(DR) in PDL- and silk-coated astrocytes is similar, indicating that silk does not alter the resting K(+) current. We also demonstrate that guanosine- (GUO) embedded silk enables the direct modulation of astroglial K(+) conductance in vitro. Astrocytes plated on GUO-embedded silk are more hyperpolarized and express inward rectifying K(+) conductance (K(ir)). The K(+) inward current increases and this is paralleled by upregulation and membrane polarization of K(ir)4.1 protein signal. Collectively these results indicate that silk is a suitable biomaterial platform for the in vitro studies of astroglial ion channel responses and related physiology.


Subject(s)
Astrocytes/drug effects , Astrocytes/physiology , Brain/cytology , Drug Delivery Systems , Electrophysiological Phenomena/drug effects , Silk/pharmacology , Animals , Bombyx/chemistry , Cell Proliferation/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Guanosine/pharmacology , Indoles/metabolism , Ion Channel Gating/drug effects , Microscopy, Confocal , Neocortex/cytology , Polylysine/pharmacology , Rats
16.
J Neurochem ; 113(3): 796-806, 2010 May.
Article in English | MEDLINE | ID: mdl-20180885

ABSTRACT

The P2X(7) receptor (P2X(7)R) is an ATP-gated cation channel whose biophysical properties remain to be unravelled unequivocally. Its activity is modulated by divalent cations and organic messengers such as arachidonic acid (AA). In this study, we analysed the differential modulation of magnesium (Mg(2+)) and AA on P2X(7)R by measuring whole-cell currents and intracellular Ca(2+) ([Ca(2+)](i)) and Na(+) ([Na(+)](i)) dynamics in HEK293 cells stably expressing full-length P2X(7)R and in cells endowed with the P2X(7)R variant lacking the entire C-terminus tail (trP2X(7)R), which is thought to control the pore activation. AA induced a robust potentiation of the P2X(7)R- and trP2X(7)R-mediated [Ca(2+)](i) rise but did not affect the ionic currents in both conditions. Extracellular Mg(2+) reduced the P2X7R- and trP2X(7)R-mediated [Ca(2+)](i) rise in a dose-dependent manner through a competitive mechanism. The modulation of the magnitude of the P2X(7)R-mediated ionic current and [Na(+)](i) rise were strongly dependent on Mg(2+) concentration but occurred in a non-competitive manner. In contrast, in cells expressing the trP2X(7)R, the small ionic currents and [Na(+)](i) signals were totally insensitive to Mg(2+). Collectively, these results support the tenet of a functional structure of P2X(7)R possessing at least two distinct conductive pathways one for Ca(2+) and another for monovalent ions, with the latter which depends on the presence of the receptor C-terminus.


Subject(s)
Neural Conduction/physiology , Receptors, Purinergic P2/physiology , Signal Transduction/physiology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Adenosine Triphosphate/physiology , Animals , Arachidonic Acid/pharmacology , Calcium Signaling/drug effects , Cell Line , Cytophotometry , Electrophysiology , Humans , Magnesium/pharmacology , Neural Conduction/drug effects , Patch-Clamp Techniques , Rats , Receptors, Purinergic P2/drug effects , Receptors, Purinergic P2X7 , Recombinant Proteins , Signal Transduction/drug effects , Sodium Channels/drug effects , Sodium Channels/physiology , Transfection
17.
Channels (Austin) ; 3(5): 323-36, 2009.
Article in English | MEDLINE | ID: mdl-19713739

ABSTRACT

Accumulating evidence indicate that the gap-junction inhibitor carbenoxolone (CBX) regulates neuronal synchronization, depresses epileptiform activity and has a neuroprotective action. These CBX effects do not depend solely on its ability to inhibit gap junction channels formed by connexins (Cx), but the underlying mechanisms remain to be elucidated. Here we addressed the questions whether CBX modulates volume-regulated anion channels (VRAC) involved in the regulatory volume decrease and regulates the associated release of excitatory amino acids in cultured rat cortical astrocytes. We found that CBX inhibits VRAC conductance with potency comparable to that able to depress the activity of the most abundant astroglial gap junction protein connexin43 (Cx43). However, the knock down of Cx43 with small interfering RNA (siRNA) oligonucleotides and the use of various pharmacological tools revealed that VRAC inhibition was not mediated by interaction of CBX with astroglial Cx proteins. Comparative experiments in HEK293 cells stably expressing another putative target of CBX, the purinergic ionotropic receptor P2X7, indicate that the presence of this receptor was not necessary for CBX-mediated depression of VRAC. Finally, we show that in COS-7 cells, which are not endowed with pannexin-1 protein, another astroglial plasma membrane interactor of CBX, VRAC current retained its sensitivity to CBX. Complementary analyses indicate that the VRAC-mediated release of excitatory amino acid aspartate was decreased by CBX. Collectively, these findings support the notion that CBX could affect astroglial ability to modulate neuronal activity by suppressing excitatory amino acid release through VRAC, thereby providing a possible mechanistic clue for the neuroprotective effect of CBX in vivo.


Subject(s)
Astrocytes/cytology , Carbenoxolone/pharmacology , Animals , Anions , Anti-Ulcer Agents/pharmacology , Astrocytes/pathology , COS Cells , Cerebral Cortex/pathology , Chlorocebus aethiops , Connexin 43/metabolism , Dose-Response Relationship, Drug , Gap Junctions , Humans , Neurons/metabolism , RNA, Small Interfering/metabolism , Rats
18.
Neurochem Int ; 53(6-8): 255-62, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18804898

ABSTRACT

Accumulating evidence indicates that astroglial syncytium plays key role in normal and pathological brain functions. Astrocytes both in vitro and in situ respond to extracellular adenine-based nucleotides via the activation of P2 receptors. Massive release of ATP from neurons and glial cells occurs as a result of pathological conditions of the brain leading to neuroinflammation and involving P2X7 receptors. In this study, we investigated whether P2X7 stimulation on cultured cortical astrocytes promoted a differential activation of mitogen-activated protein kinases (MAPKs), and whether the second messenger arachidonic acid (AA), which is also a key modulator of neuroinflammation, affected the P2X7-mediated MAPK phosphorylation. The results show that the synthetic P2X7 receptor agonist 2',3'-O-(4-benzoyl)benzoyl-ATP (BzATP), induced a concentration-dependent phosphorylation of MAPK ERK1/2, JNK and p38. Stimulation of ERK1/2, JNK and p38 phosphorylation was also obtained by pathophysiological levels of extracellularly applied AA. Interestingly, a robust potentiation of ERK1/2 phosphorylation was elicited by co-application of BzATP and AA, whereas no differences were observed in JNK or p38 phosphosignals. The kinases activation showed a differential dependence on the presence of extracellular Ca(2+). The potentiation of BzATP-mediated ERK1/2 phosphorylation was also observed in human embryonic kidney cells (HEK293) stably transfected with rat P2X7, but not in HEK cells expressing truncated P2X7 receptor lacking the full cytoplasmic carboxy-terminal or in those carrying the structurally related rat P2X2. AA and BzATP synergism in ERK1/2 activation was abolished by cyclo-oxygenase and lipoxygenase pathway inhibitors. The result that ERK1/2-mediated transduction pathway is synergistically modulated by ATP and AA signalling depicts possible novel pharmacological targets for interfering with pathological activation of astroglial cells.


Subject(s)
Arachidonic Acid/metabolism , Astrocytes/metabolism , Encephalitis/metabolism , MAP Kinase Signaling System/physiology , Receptor Cross-Talk/physiology , Receptors, Purinergic P2/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Affinity Labels/pharmacology , Animals , Animals, Newborn , Cell Line , Cells, Cultured , Dose-Response Relationship, Drug , Drug Synergism , Encephalitis/physiopathology , Humans , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Purinergic P2 Receptor Agonists , Rats , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X7 , Transfection
19.
J Neurochem ; 100(1): 87-104, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17064359

ABSTRACT

In the brain, the astroglial syncytium is crucially involved in the regulation of water homeostasis. Accumulating evidence indicates that a dysregulation of the astrocytic processes controlling water homeostasis has a pathogenetic role in several brain injuries. Here, we have analysed by RNA interference technology the functional interactions occurring between the most abundant water channel in the brain, aquaporin-4 (AQP4), and the swelling-activated Cl(-) current expressed by cultured rat cortical astrocytes. We show that in primary cultured rat cortical astrocytes transfected with control small interfering RNA (siRNA), hypotonic shock promotes an increase in cellular volume accompanied by augmented membrane conductance mediated by volume-regulated anion channels (VRAC). Conversely, astroglia in which AQP4 was knocked down (AQP4 KD) by transfection with AQP4 siRNA changed their morphology from polygonal to process-bearing, and displayed normal cell swelling but reduced VRAC activity. Pharmacological manipulations of actin cytoskeleton in rat astrocytes, and functional analysis in mouse astroglial cells, which retain their morphology upon knockdown of AQP4, suggest that stellation of AQP4 KD rat cortical astrocytes was not causally linked to reduction of VRAC current. Molecular analysis of possible candidates of swelling-activated Cl(-) current provided evidence that in AQP4 KD astrocytes, there was a down-regulation of chloride channel-2 (CIC-2), which, however, was not involved in VRAC conductance. Inclusion of ATP in the intracellular saline restored VRAC activity upon hypotonicity. Collectively, these results support the view that in cultured astroglial cells, plasma membrane proteins involved in cell volume homeostasis are assembled in a functional platform.


Subject(s)
Aquaporin 4/physiology , Astrocytes/metabolism , Cerebral Cortex/cytology , Down-Regulation/physiology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , Animals, Newborn , Aquaporin 4/genetics , Astrocytes/drug effects , Blotting, Western/methods , Bucladesine/pharmacology , CLC-2 Chloride Channels , Cells, Cultured , Chloride Channels/physiology , Dose-Response Relationship, Radiation , Down-Regulation/drug effects , Electric Stimulation/methods , Fluorescent Antibody Technique , Immunoprecipitation/methods , Ion Channel Gating/physiology , Isotonic Solutions/pharmacology , Membrane Potentials/physiology , Mice , Patch-Clamp Techniques , Phloretin/pharmacology , RNA, Small Interfering/metabolism , Rats , Transfection/methods
20.
J Pain ; 7(10): 735-46, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17018334

ABSTRACT

UNLABELLED: Vanilloid receptor subunit 1 (TRPV1) is an integrator of physical and chemical stimuli in the peripheral nervous system. This receptor plays a key role in the pathophysiology of inflammatory pain. Thus, the identification of receptor antagonists with analgesic and anti-inflammatory activity in vivo is an important goal of current neuropharmacology. Here, we report that [L-arginyl]-[N-[2,4-dichlorophenethyl]glycyl]-N-(2,4-dichlorophenethyl) glycinamide (H-Arg-15-15C) is a channel blocker that abrogates capsaicin and pH-evoked TRPV1 channel activity with submicromolar activity. Compound H-Arg-15-15C preferentially inhibits TRPV1, showing marginal block of other neuronal receptors. Compound H-Arg-15-15C acts as a noncompetitive capsaicin antagonist with modest voltage-dependent blockade activity. The compound inhibited capsaicin-evoked nerve activity in afferent fibers without affecting mechanically activated activity. Notably, administration of compound H-Arg-15-15C prevented the irritant activity of a local administration of capsaicin and formalin and reversed the thermal hyperalgesia evoked by injection of complete Freund's adjuvant. Furthermore, it attenuated carrageenan-induced paw inflammation. Compound H-Arg-15-15C specifically decreased inflammatory conditions without affecting normal nociception. Taken together, these findings demonstrate that compound H-Arg-15-15C is a channel blocker of TRPV1 with analgesic and anti-inflammatory activity in vivo at clinically useful doses and substantiate the tenet that TRPV1 plays an important role in the etiology of chronic inflammatory pain. PERSPECTIVE: This study reports the design of a potent TRPV1 noncompetitive antagonist that exhibits anti-inflammatory and analgesic activity in preclinical models of acute and chronic pain. This compound is a lead for analgesic drug development.


Subject(s)
Analgesics/pharmacology , Anti-Inflammatory Agents/pharmacology , Arginine/analogs & derivatives , Glycine/analogs & derivatives , Ion Channels/antagonists & inhibitors , Nociceptors/drug effects , Pain/drug therapy , TRPV Cation Channels/antagonists & inhibitors , Analgesics/chemical synthesis , Analgesics/chemistry , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/chemistry , Arginine/chemical synthesis , Arginine/chemistry , Arginine/pharmacology , COS Cells , Capsaicin/antagonists & inhibitors , Chlorocebus aethiops , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Design , Female , Glycine/chemical synthesis , Glycine/chemistry , Glycine/pharmacology , Inflammation/drug therapy , Inflammation/physiopathology , Ion Channels/metabolism , Male , Mice , Mice, Inbred ICR , Nociceptors/metabolism , Nociceptors/physiopathology , Oocytes , Pain/physiopathology , Patch-Clamp Techniques , Rats , Rats, Wistar , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , TRPV Cation Channels/metabolism , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...