Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nat Neurosci ; 21(10): 1412-1420, 2018 10.
Article in English | MEDLINE | ID: mdl-30224804

ABSTRACT

Schizophrenia is a severely debilitating neurodevelopmental disorder. Establishing a causal link between circuit dysfunction and particular behavioral traits that are relevant to schizophrenia is crucial to shed new light on the mechanisms underlying the pathology. We studied an animal model of the human 22q11 deletion syndrome, the mutation that represents the highest genetic risk of developing schizophrenia. We observed a desynchronization of hippocampal neuronal assemblies that resulted from parvalbumin interneuron hypoexcitability. Rescuing parvalbumin interneuron excitability with pharmacological or chemogenetic approaches was sufficient to restore wild-type-like CA1 network dynamics and hippocampal-dependent behavior during adulthood. In conclusion, our data provide insights into the network dysfunction underlying schizophrenia and highlight the use of reverse engineering to restore physiological and behavioral phenotypes in an animal model of neurodevelopmental disorder.


Subject(s)
CA1 Region, Hippocampal/pathology , Mental Disorders/etiology , Nerve Net/pathology , Nonlinear Dynamics , Schizophrenia/pathology , Schizophrenia/physiopathology , 22q11 Deletion Syndrome/complications , 22q11 Deletion Syndrome/genetics , Action Potentials/drug effects , Action Potentials/physiology , Animals , Animals, Newborn , Clozapine/analogs & derivatives , Clozapine/pharmacology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/physiopathology , Neuregulins/pharmacology , Neurons/drug effects , Neurons/physiology , Parvalbumins/genetics , Parvalbumins/metabolism , Prepulse Inhibition/physiology , Reflex, Startle/physiology , Schizophrenia/etiology , Schizophrenia/genetics
2.
Proc Natl Acad Sci U S A ; 113(11): 3060-5, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26929355

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease affecting motor neurons that leads to progressive paralysis of skeletal muscle. Studies of ALS have revealed defects in expression of acetylcholine receptors (AChRs) in skeletal muscle that occur even in the absence of motor neuron anomalies. The endocannabinoid palmitoylethanolamide (PEA) modified the clinical conditions in one ALS patient, improving muscle force and respiratory efficacy. By microtransplanting muscle membranes from selected ALS patients into Xenopus oocytes, we show that PEA reduces the desensitization of acetylcholine-evoked currents after repetitive neurotransmitter application (i.e., rundown). The same effect was observed using muscle samples from denervated (non-ALS) control patients. The expression of human recombinant α1ß1γδ (γ-AChRs) and α1ß1εδ AChRs (ε-AChRs) in Xenopus oocytes revealed that PEA selectively affected the rundown of ACh currents in ε-AChRs. A clear up-regulation of the α1 subunit in muscle from ALS patients compared with that from non-ALS patients was found by quantitative PCR, but no differential expression was found for other subunits. Clinically, ALS patients treated with PEA showed a lower decrease in their forced vital capacity (FVC) over time as compared with untreated ALS patients, suggesting that PEA can enhance pulmonary function in ALS. In the present work, data were collected from a cohort of 76 ALS patients and 17 denervated patients. Our results strengthen the evidence for the role of skeletal muscle in ALS pathogenesis and pave the way for the development of new drugs to hamper the clinical effects of the disease.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Ethanolamines/therapeutic use , Molecular Targeted Therapy , Muscle, Skeletal/drug effects , Palmitic Acids/therapeutic use , Receptors, Nicotinic/drug effects , Action Potentials/drug effects , Adult , Aged , Aged, 80 and over , Amides , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Cell Membrane/drug effects , Cell Membrane/transplantation , Ethanolamines/pharmacology , Female , Humans , Male , Microinjections , Middle Aged , Muscle Denervation , Muscle, Skeletal/ultrastructure , Neuromuscular Junction/physiopathology , Oocytes , Palmitic Acids/pharmacology , Receptors, Nicotinic/physiology , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/genetics , Single-Blind Method , Xenopus laevis
3.
Neuron ; 89(5): 1074-85, 2016 Mar 02.
Article in English | MEDLINE | ID: mdl-26875623

ABSTRACT

Hippocampal neurons activated during encoding drive the recall of contextual fear memory. Little is known about how such ensembles emerge during acquisition and eventually form the cellular engram. Manipulating the activity of granule cells (GCs) of the dentate gyrus (DG), we reveal a mechanism of lateral inhibition that modulates the size of the cellular engram. GCs engage somatostatin-positive interneurons that inhibit the dendrites of surrounding GCs. Our findings reveal a microcircuit within the DG that controls the size of the cellular engram and the stability of contextual fear memory.


Subject(s)
Cell Size , Dentate Gyrus/cytology , Interneurons/physiology , Memory/physiology , Somatostatin/metabolism , Action Potentials/genetics , Animals , Cell Count , Channelrhodopsins , Conditioning, Psychological/physiology , Fear/physiology , Luminescent Proteins/genetics , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Parvalbumins/genetics , Parvalbumins/metabolism , Patch-Clamp Techniques , Somatostatin/genetics , Spatial Behavior/physiology , Statistics, Nonparametric
4.
Front Cell Neurosci ; 9: 409, 2015.
Article in English | MEDLINE | ID: mdl-26528137

ABSTRACT

Adenosine is a widespread neuromodulator within the CNS and its extracellular level is increased during hypoxia or intense synaptic activity, modulating pre- and postsynaptic sites. We studied the neuromodulatory action of adenosine on glutamatergic currents in the hippocampus, showing that activation of multiple adenosine receptors (ARs) by basal adenosine impacts postsynaptic site. Specifically, the stimulation of both A1R and A3R reduces AMPA currents, while A2AR has an opposite potentiating effect. The effect of ARs stimulation on glutamatergic currents in hippocampal cultures was investigated using pharmacological and genetic approaches. A3R inhibition by MRS1523 increased GluR1-Ser845 phosphorylation and potentiated AMPA current amplitude, increasing the apparent affinity for the agonist. A similar effect was observed blocking A1R with DPCPX or by genetic deletion of either A3R or A1R. Conversely, impairment of A2AR reduced AMPA currents, and decreased agonist sensitivity. Consistently, in hippocampal slices, ARs activation by AR agonist NECA modulated glutamatergic current amplitude evoked by AMPA application or afferent fiber stimulation. Opposite effects of AR subtypes stimulation are likely associated to changes in GluR1 phosphorylation and represent a novel mechanism of physiological modulation of glutamatergic transmission by adenosine, likely acting in normal conditions in the brain, depending on the level of extracellular adenosine and the distribution of AR subtypes.

5.
Neurobiol Dis ; 82: 311-320, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26168875

ABSTRACT

Temporal lobe epilepsy (TLE) is the most prevalent form of adult focal onset epilepsy often associated with drug-resistant seizures. Numerous studies suggest that neuroinflammatory processes are pathologic hallmarks of both experimental and human epilepsy. In particular, the interleukin (IL)-1ß/IL-1 receptor type 1 (R1) axis is activated in epileptogenic tissue, where it contributes significantly to the generation and recurrence of seizures in animal models. In this study, we investigated whether IL-1ß affects the GABA-evoked currents (I(GABA)) in TLE tissue from humans. Given the limited availability of fresh human brain specimens, we used the "microtransplantation" method of injecting Xenopus oocytes with membranes from surgically resected hippocampal and cortical tissue from 21 patients with TLE and hippocampal sclerosis (HS), hippocampal tissue from five patients with TLE without HS, and autoptic and surgical brain specimens from 15 controls without epilepsy. We report the novel finding that pathophysiological concentrations of IL-1ß decreased the I(GABA) amplitude by up to 30% in specimens from patients with TLE with or without HS, but not in control tissues. This effect was reproduced by patch-clamp recordings on neurons in entorhinal cortex slices from rats with chronic epilepsy, and was not observed in control slices. In TLE specimens from humans, the IL-1ß effect was mediated by IL-1R1 and PKC. We also showed that IL-1R1 and IRAK1, the proximal kinase mediating the IL-1R1 signaling, are both up-regulated in the TLE compared with control specimens, thus supporting the idea that the IL-1ß/IL-R1 axis is activated in human epilepsy. Our findings suggest a novel mechanism possibly underlying the ictogenic action of IL-1ß, thus suggesting that this cytokine contributes to seizure generation in human TLE by reducing GABA-mediated neurotransmission.


Subject(s)
Cerebral Cortex/physiopathology , Epilepsy, Temporal Lobe/physiopathology , Hippocampus/physiopathology , Interleukin-1beta/metabolism , Receptors, GABA-A/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cerebral Cortex/pathology , Cerebral Cortex/surgery , Disease Models, Animal , Epilepsy, Temporal Lobe/pathology , Epilepsy, Temporal Lobe/surgery , Female , GABA Agents/administration & dosage , Hippocampus/pathology , Hippocampus/surgery , Humans , Interleukin-1beta/administration & dosage , Kainic Acid , Male , Middle Aged , Oocytes , Patch-Clamp Techniques , Rats, Sprague-Dawley , Tissue Culture Techniques , Transplantation, Heterologous/methods , Xenopus , Young Adult
6.
Epilepsia ; 54(10): 1834-44, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24032743

ABSTRACT

PURPOSE: The chemokine fractalkine/CX3CL1 and its receptor CX3CR1 are widely expressed in the central nervous system (CNS). Recent evidence showed that CX3CL1 participates in inflammatory responses that are common features of CNS disorders, such as epilepsy. Mesial temporal lobe epilepsy (MTLE) is the prevalent form of focal epilepsy in adults, and hippocampal sclerosis (HS) represents the most common underlying pathologic abnormality, as demonstrated at autopsy and postresection studies. Relevant features of MTLE are a characteristic pattern of neuronal loss, as are astrogliosis and microglia activation. Several factors affect epileptogenesis in patients with MTLE, including a lack of γ-aminobutyric acid (GABA)ergic inhibitory efficacy. Therefore, experiments were designed to investigate whether, in MTLE brain tissues, CX3CL1 may influence GABAA receptor (GABAA R) mediated transmission, with a particular focus on the action of CX3CL1 on the use-dependent decrease (rundown) of the GABA-evoked currents (IGABA ), a feature underlying the reduction of GABAergic function in epileptic tissue. METHODS: Patch-clamp recordings were obtained from cortical pyramidal neurons in slices from six MTLE patients after surgery. Alternatively, the cell membranes from epileptic brain tissues of 17 MTLE patients or from surgical samples and autopsies of nonepileptic patients were microtransplanted into Xenopus oocytes, and IGABA were recorded using the standard two-microelectrode voltage-clamp technique. Immunohistochemical staining and double-labeling studies were carried out on the same brain tissues to analyze CX3CR1 expression. KEY FINDINGS: In native pyramidal neurons from cortical slices of patients with MTLE, CX3CL1 reduced IGABA rundown and affected the recovery of IGABA amplitude from rundown. These same effects were confirmed in oocytes injected with cortical and hippocampal MTLE membranes, whereas CX3CL1 did not influence IGABA in oocytes injected with nonepileptic tissues. Consistent with a specific effect of CX3CL1 on tissues from patients with MTLE, CX3CR1 immunoreactivity was higher in MTLE sclerotic hippocampi than in control tissues, with a prominent expression in activated microglial cells. SIGNIFICANCE: These findings indicate a role for CX3CL1 in MTLE, supporting recent evidence on the relevance of brain inflammation in human epilepsies. Our data demonstrate that in MTLE tissues the reduced GABAergic function can be modulated by CX3CL1. The increased CX3CR1 expression in microglia and the modulation by CX3CL1 of GABAergic currents in human epileptic brain suggests new therapeutic approaches for drug-resistant epilepsies based on the evidence that the propagation of seizures can be influenced by inflammatory processes.


Subject(s)
Chemokine CX3CL1/physiology , Epilepsy, Temporal Lobe/physiopathology , Receptors, GABA-A/physiology , Action Potentials/physiology , Adult , Animals , Blotting, Western , Brain/physiopathology , Cell Membrane/physiology , Female , Humans , Male , Middle Aged , Oocytes/physiology , Pyramidal Cells/physiology , Xenopus laevis , Young Adult
7.
Front Mol Neurosci ; 5: 100, 2012.
Article in English | MEDLINE | ID: mdl-23189038

ABSTRACT

Neuronal chloride concentration ([Cl(-)](i)) is known to be dynamically modulated and alterations in Cl(-) homeostasis may occur in the brain at physiological and pathological conditions, being also likely involved in glioma-related seizures. However, the mechanism leading to changes in neuronal [Cl(-)](i) during glioma invasion are still unclear. To characterize the potential effect of glioma released soluble factors on neuronal [Cl(-)](i), we used genetically encoded CFP/YFP-based ratiometric Cl-(apical) Sensor transiently expressed in cultured hippocampal neurons. Exposition of neurons to glioma conditioned medium (GCM) caused rapid and transient elevation of [Cl(-)](i), resulting in the increase of fluorescence ratio, which was strongly reduced by blockers of ionotropic glutamate receptors APV and NBQX. Furthermore, in HEK cells expressing GluR1-AMPA receptors, GCM activated ionic currents with efficacy similar to those caused by glutamate, supporting the notion that GCM contains glutamate or glutamatergic agonists, which cause neuronal depolarization, activation of NMDA and AMPA/KA receptors leading to elevation of [Cl(-)](i). Chromatographic analysis of the GCM showed that it contained several aminoacids, including glutamate, whose release from glioma cells did not occur via the most common glial mechanisms of transport, or in response to hypoosmotic stress. GCM also contained glycine, whose action contrasted the glutamate effect. Indeed, strychnine application significantly increased GCM-induced depolarization and [Cl(-)](i) rise. GCM-evoked [Cl(-)](i) elevation was not inhibited by antagonists of Cl(-) transporters and significantly reduced in the presence of anion channels blocker NPPB, suggesting that Cl(-) selective channels are a major route for GCM-induced Cl(-) influx. Altogether, these data show that glioma released aminoacids may dynamically alter Cl(-) equilibrium in surrounding neurons, deeply interfering with their inhibitory balance, likely leading to physiological and pathological consequences.

8.
Neuron ; 67(4): 656-66, 2010 Aug 26.
Article in English | MEDLINE | ID: mdl-20797541

ABSTRACT

The central nucleus of the amygdala (CeA) serves as a major output of this structure and plays a critical role in the expression of conditioned fear. By combining cell- and tissue-specific pharmacogenetic inhibition with functional magnetic resonance imaging (fMRI), we identified circuits downstream of CeA that control fear expression in mice. Selective inhibition of a subset of neurons in CeA led to decreased conditioned freezing behavior and increased cortical arousal as visualized by fMRI. Correlation analysis of fMRI signals identified functional connectivity between CeA, cholinergic forebrain nuclei, and activated cortical structures, and cortical arousal was blocked by cholinergic antagonists. Importantly, inhibition of these neurons switched behavioral responses to the fear stimulus from passive to active responses. Our findings identify a neural circuit in CeA that biases fear responses toward either passive or active coping strategies.


Subject(s)
Amygdala/physiology , Brain/physiology , Fear/physiology , Neurons/physiology , Action Potentials/drug effects , Action Potentials/physiology , Adaptation, Psychological/drug effects , Adaptation, Psychological/physiology , Amygdala/blood supply , Amygdala/drug effects , Animals , Blood Volume/drug effects , Brain/blood supply , Brain/drug effects , Cerebrovascular Circulation/drug effects , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Fear/drug effects , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , In Vitro Techniques , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Models, Neurological , Neural Pathways/blood supply , Neural Pathways/drug effects , Neural Pathways/physiology , Neurons/drug effects , Oxytocin/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin 5-HT1 Receptor Agonists , Serotonin 5-HT1 Receptor Antagonists
9.
Am J Physiol Cell Physiol ; 299(1): C175-84, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20392929

ABSTRACT

The activation of ion channels is crucial during cell movement, including glioblastoma cell invasion in the brain parenchyma. In this context, we describe for the first time the contribution of intermediate conductance Ca(2+)-activated K (IK(Ca)) channel activity in the chemotactic response of human glioblastoma cell lines, primary cultures, and freshly dissociated tissues to CXC chemokine ligand 12 (CXCL12), a chemokine whose expression in glioblastoma has been correlated with its invasive capacity. We show that blockade of the IK(Ca) channel with its specific inhibitor 1-[(2-chlorophenyl) diphenylmethyl]-1H-pyrazole (TRAM-34) or IK(Ca) channel silencing by short hairpin RNA (shRNA) completely abolished CXCL12-induced cell migration. We further demonstrate that this is not a general mechanism in glioblastoma cell migration since epidermal growth factor (EGF), which also activates IK(Ca) channels in the glioblastoma-derived cell line GL15, stimulate cell chemotaxis even if the IK(Ca) channels have been blocked or silenced. Furthermore, we demonstrate that both CXCL12 and EGF induce Ca(2+) mobilization and IK(Ca) channel activation but only CXCL12 induces a long-term upregulation of the IK(Ca) channel activity. Furthermore, the Ca(2+)-chelating agent BAPTA-AM abolished the CXCL12-induced, but not the EGF-induced, glioblastoma cell chemotaxis. In addition, we demonstrate that the extracellular signal-regulated kinase (ERK)1/2 pathway is only partially implicated in the modulation of CXCL12-induced glioblastoma cell movement, whereas the phosphoinositol-3 kinase (PI3K) pathway is not involved. In contrast, EGF-induced glioblastoma migration requires both ERK1/2 and PI3K activity. All together these findings suggest that the efficacy of glioblastoma invasiveness might be related to an array of nonoverlapping mechanisms activated by different chemotactic agents.


Subject(s)
Brain Neoplasms/metabolism , Chemokine CXCL12/metabolism , Chemotaxis , Glioblastoma/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Brain Neoplasms/pathology , Calcium Signaling , Cell Line, Tumor , Chelating Agents/pharmacology , Epidermal Growth Factor/metabolism , Glioblastoma/pathology , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Membrane Potentials , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Potassium Channel Blockers/pharmacology , Protein Kinase Inhibitors/pharmacology , RNA Interference , Receptors, CXCR4/metabolism , Recombinant Proteins/metabolism , Tumor Cells, Cultured
10.
J Neuroimmunol ; 215(1-2): 36-42, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19709758

ABSTRACT

We have examined how the chemokine fractalkine/CX(3)CL1 influences long-term potentiation (LTP) in CA1 mouse hippocampal slices. Field potentials (fEPSPs) were recorded upon electrical stimulation of Schaffer collaterals. It was found that application of CX(3)CL1 inhibits LTP when present during the critical induction period. LTP impairment (i) failed to occur in CX(3)CR1 deficient mice (CX(3)CR1(GFP/GFP)) and in the presence of okadaic acid (OA); (ii) required the activation of adenosine receptor 3 (A(3)R), since it was prevented in A(3)R-deficient mice or by MRS1523, a selective A(3)R antagonist. Together, these findings indicate that CX(3)CL1 inhibits hippocampal LTP through A(3)R activity.


Subject(s)
Chemokine CX3CL1/physiology , Hippocampus/immunology , Long-Term Potentiation/immunology , Receptor, Adenosine A3/metabolism , Animals , Hippocampus/metabolism , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Inhibition/immunology , Receptor, Adenosine A3/physiology
11.
J Neurosci ; 26(41): 10488-98, 2006 Oct 11.
Article in English | MEDLINE | ID: mdl-17035533

ABSTRACT

We examined the effects of the chemokine fractalkine (CX3CL1) on EPSCs evoked by electrical stimulation of Schaffer collaterals in patch-clamped CA1 pyramidal neurons from rat hippocampal slices. Acute application of CX3CL1 caused a sustained reduction of EPSC amplitude, with partial recovery after washout. CX3CL1-induced EPSC depression is postsynaptic in nature, because paired-pulse ratio was maintained, amplitude distribution of spontaneous excitatory postsynaptic currents shifted to lower values, and whole-cell current responses to AMPA were reversibly inhibited. EPSC depression by CX3CL1 is mediated by CX3CL1 receptor (CX3CR1), because CX3CL1 was unable to influence EPSC amplitude in CA1 pyramidal neurons from CX3CR1 knock-out mice. CX3CL1-induced depression of both EPSC and AMPA current was not observed in the absence of afferent fiber stimulation or AMPA receptor activation, respectively, indicating the requirement of sustained receptor activity for its development. Findings obtained from hippocampal slices, cultured hippocampal neurons, and transfected human embryonic kidney cells indicate that a Ca2+-, cAMP-, and phosphatase-dependent process is likely to modulate CX3CL1 effects because of the following: (1) CX3CL1-induced depression was antagonized by intracellular BAPTA, 8Br-cAMP, phosphatase inhibitors, and pertussis toxin (PTX); (2) CX3CL1 inhibited forskolin-induced cAMP formation sensitive to PTX; and (3) CX3CL1 inhibited forskolin-induced Ser845 GluR1 phosphorylation, which was sensitive to PTX and dependent on Ca2+ and phosphatase activity. Together, these findings indicate that CX3CL1 negatively modulates AMPA receptor function at active glutamatergic synapses through cell-signaling pathways by influencing the balance between kinase and phosphatase activity.


Subject(s)
Chemokines, CX3C/metabolism , Glutamic Acid/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Synapses/metabolism , Animals , CX3C Chemokine Receptor 1 , Cell Line , Cells, Cultured , Chemokine CX3CL1 , Chemokines, CX3C/genetics , Chemokines, CX3C/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Membrane Proteins/genetics , Membrane Proteins/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Synapses/drug effects
12.
Neuropharmacology ; 51(4): 816-21, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16815480

ABSTRACT

This work reports the effect of chemokine fractalkine/CX3CL1, an endogenous small peptide highly expressed in the central nervous system, on evoked synaptic responses investigated in mouse CA1 stratum radiatum using an electrophysiological approach. We report that in acute mouse hippocampal slices, superfusion of CX3CL1 resulted in a reversible depression of the field excitatory postsynaptic potential (fEPSP) which developed within few seconds, increased for up to 10 min of application and disappeared within 30 min after the end of CX3CL1 treatment. We also show that CX3CL1-induced synaptic depression is (i) dose-dependent with IC50 and nH values of 0.7 nM and 1, respectively, (ii) not associated with a change in paired-pulse facilitation, (iii) mediated through CX3CL1 receptor (CX3CR1), being absent in CX3CR1-/- mice and inhibited in wild-type mice by a specific blocking antibody, and (iv) occluded by the induction of homosynaptic long-term depression (LTD). We conclude that CX3CL1 is a potent neuromodulator of the evoked excitatory synaptic transmission, sharing common mechanisms with LTD.


Subject(s)
Excitatory Postsynaptic Potentials/physiology , Hippocampus/physiology , Receptors, Chemokine/physiology , Synaptic Transmission/physiology , Animals , Animals, Newborn , CX3C Chemokine Receptor 1 , Chemokine CX3CL1 , Chemokines, CX3C/immunology , Chemokines, CX3C/pharmacology , Dose-Response Relationship, Drug , Electric Stimulation/methods , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/radiation effects , Hippocampus/cytology , Hippocampus/drug effects , Humans , In Vitro Techniques , Membrane Proteins/immunology , Membrane Proteins/pharmacology , Mice , Mice, Knockout , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neural Inhibition/radiation effects , Patch-Clamp Techniques/methods , Receptors, Chemokine/deficiency , Synaptic Transmission/drug effects , Synaptic Transmission/radiation effects
13.
J Neuroimmunol ; 166(1-2): 19-28, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16019082

ABSTRACT

Excitotoxicity is a cell death caused by excessive exposure to glutamate (Glu), contributing to neuronal degeneration in many acute and chronic CNS diseases. We explored the role of fractalkine/CX3CL1 on survival of hippocampal neurons exposed to excitotoxic doses of Glu. We found that: CX3CL1 reduces excitotoxicity when co-applied with Glu, through the activation of the ERK1/2 and PI3K/Akt pathways, or administered up to 8 h after Glu insult; CX3CL1 reduces the Glu-activated whole-cell current through mechanisms dependent on intracellular Ca2+; CX3CL1 is released from hippocampal cells after excitotoxic insult, likely providing an endogenous protective mechanism against excitotoxic cell death.


Subject(s)
Chemokines, CX3C/physiology , Glutamic Acid/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Membrane Proteins/physiology , Neuroprotective Agents , Neurotoxins/pharmacology , Animals , Cell Survival/physiology , Cells, Cultured , Chemokine CX3CL1 , Chemokines, CX3C/administration & dosage , Chemokines, CX3C/metabolism , Chemokines, CX3C/pharmacology , Drug Administration Schedule , Drug Combinations , Electric Conductivity , Excitatory Amino Acid Agonists/pharmacology , Hippocampus/physiology , Membrane Proteins/administration & dosage , Membrane Proteins/metabolism , Membrane Proteins/pharmacology , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Rats , Rats, Wistar , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...