Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Glia ; 72(6): 1096-1116, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38482984

ABSTRACT

The medial prefrontal cortex (mPFC) is involved in cognitive functions such as working memory. Astrocytic cannabinoid type 1 receptor (CB1R) induces cytosolic calcium (Ca2+) concentration changes with an impact on neuronal function. mPFC astrocytes also express adenosine A1 and A2A receptors (A1R, A2AR), being unknown the crosstalk between CB1R and adenosine receptors in these cells. We show here that a further level of regulation of astrocyte Ca2+ signaling occurs through CB1R-A2AR or CB1R-A1R heteromers that ultimately impact mPFC synaptic plasticity. CB1R-mediated Ca2+ transients increased and decreased when A1R and A2AR were activated, respectively, unveiling adenosine receptors as modulators of astrocytic CB1R. CB1R activation leads to an enhancement of long-term potentiation (LTP) in the mPFC, under the control of A1R but not of A2AR. Notably, in IP3R2KO mice, that do not show astrocytic Ca2+ level elevations, CB1R activation decreases LTP, which is not modified by A1R or A2AR. The present work suggests that CB1R has a homeostatic role on mPFC LTP, under the control of A1R, probably due to physical crosstalk between these receptors in astrocytes that ultimately alters CB1R Ca2+ signaling.


Subject(s)
Astrocytes , Cannabinoids , Mice , Animals , Receptors, Cannabinoid , Receptor, Adenosine A2A , Neuronal Plasticity , Receptor, Cannabinoid, CB1/genetics
2.
bioRxiv ; 2023 Jul 19.
Article in English | MEDLINE | ID: mdl-37503130

ABSTRACT

Astrocytes are active cells involved in brain function through the bidirectional communication with neurons, in which the astrocyte calcium signal plays a crucial role. Synaptically-evoked calcium increases can be localized to independent subcellular domains or expand to the entire cell, i.e., calcium surge. In turn, astrocytes may regulate individual synapses by calcium-dependent release of gliotransmitters. Because a single astrocyte may contact ~100,000 synapses, the control of the intracellular calcium signal propagation may have relevant consequences on brain function by regulating the spatial range of astrocyte neuromodulation of synapses. Yet, the properties governing the spatial dynamics of the astrocyte calcium signal remains poorly defined. Imaging subcellular responses of cortical astrocytes to sensory stimulation in mice, we show that sensory-evoked astrocyte calcium responses originated and remained localized in domains of the astrocytic arborization, but eventually propagated to the entire cell if a spatial threshold of >23% of the arborization being activated was surpassed. Using transgenic IP3R2-/- mice, we found that type-2 IP3 receptors were necessary for the generation of the astrocyte calcium surge. We finally show using in situ electrophysiological recordings that the spatial threshold of the astrocyte calcium signal consequently determined the gliotransmitter release. Present results reveal a fundamental property of astrocyte calcium physiology, i.e., a spatial threshold for the astrocyte intracellular calcium signal propagation, which depends on astrocyte intrinsic properties and governs the astrocyte integration of local synaptic activity and the subsequent neuromodulation.

3.
Cells ; 12(9)2023 04 26.
Article in English | MEDLINE | ID: mdl-37174653

ABSTRACT

While neurons have traditionally been considered the primary players in information processing, the role of astrocytes in this mechanism has largely been overlooked due to experimental constraints. In this review, we propose that astrocytic ensembles are active working groups that contribute significantly to animal conduct and suggest that studying the maps of these ensembles in conjunction with neurons is crucial for a more comprehensive understanding of behavior. We also discuss available methods for studying astrocytes and argue that these ensembles, complementarily with neurons, code and integrate complex behaviors, potentially specializing in concrete functions.


Subject(s)
Astrocytes , Neurons , Animals , Astrocytes/physiology , Neurons/physiology , Cognition , Models, Neurological
4.
Cancer ; 129(5): 771-779, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36504077

ABSTRACT

BACKGROUND: Children with relapsed acute lymphoblastic leukemia (ALL) in low-income and middle-income countries rarely survive. The Pediatric Hematology-Oncology Association of Central America (AHOPCA) developed the AHOPCA-ALL REC 2014 protocol to improve outcomes in resource-constrained settings without access to stem cell transplantation. METHODS: The AHOPCA-ALL REC 2014 protocol was based on a modified frontline induction phase 1A, a consolidation therapy with six modified R-blocks derived from the ALL-Berlin-Frankfurt-Munster REZ 2002 protocol and intermittent maintenance therapy. Children with B-lineage ALL were eligible after a late medullary relapse, an early or late combined relapse, or any extramedullary relapses. Those with T-lineage ALL were eligible after early and late extramedullary relapses, as were those with both B-lineage and T-lineage relapses occurring at least 3 months after therapy abandonment. RESULTS: The study population included 190 patients with T-lineage (n = 3) and B-lineage (n = 187) ALL. Of those with B-lineage ALL, 25 patients had a very early extramedullary relapse, 40 had an early relapse (32 extramedullary and 8 combined), and 125 had a late relapse (34 extramedullary, 19 combined, and 72 medullary). The main cause of treatment failure was second relapse (52.1%). The 3-year event-free survival rate (± standard error) was 25.9% ± 3.5%, and the 3-year overall survival rate was 36.7% ± 3.8%. The 3-year event-free survival rate was 47.2% ± 4.7% for late relapses. The most frequently reported toxicity was grade 3 or 4 infection. Mortality during treatment occurred in 17 patients (8.9%), in most cases because of infectious complications. CONCLUSIONS: Selected children with relapsed ALL in Central America can be cured with second-line regimens even without access to consolidation with stem cell transplantation. Children in low-income and middle-income countries who have lower risk relapses of ALL should be treated with curative intent.


Subject(s)
Hematopoietic Stem Cell Transplantation , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Child , Humans , Developing Countries , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Recurrence , Antineoplastic Combined Chemotherapy Protocols , Poverty
5.
Nat Commun ; 13(1): 5272, 2022 09 07.
Article in English | MEDLINE | ID: mdl-36071061

ABSTRACT

Astrocytes are key elements of brain circuits that are involved in different aspects of the neuronal physiology relevant to brain functions. Although much effort is being made to understand how the biology of astrocytes affects brain circuits, astrocytic network heterogeneity and plasticity is still poorly defined. Here, we have combined structural and functional imaging of astrocyte activity recorded in mice using the Ca2+-modulated photoactivatable ratiometric integrator and specific optostimulation of glutamatergic pathways to map the functional neuron-astrocyte circuitries in the nucleus accumbens (NAc). We showed pathway-specific astrocytic responses induced by selective optostimulation of main inputs from the prefrontal cortex, basolateral amygdala, and ventral hippocampus. Furthermore, co-stimulation of glutamatergic pathways induced non-linear Ca2+-signaling integration, revealing integrative properties of NAc astrocytes. All these results demonstrate the existence of specific neuron-astrocyte circuits in the NAc, providing an insight to the understanding of how the NAc integrates information.


Subject(s)
Basolateral Nuclear Complex , Nucleus Accumbens , Animals , Astrocytes/metabolism , Hippocampus/physiology , Mice , Neurons/metabolism , Nucleus Accumbens/metabolism
6.
Proc Natl Acad Sci U S A ; 119(29): e2204527119, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858325

ABSTRACT

Mice with insulin receptor (IR)-deficient astrocytes (GFAP-IR knockout [KO] mice) show blunted responses to insulin and reduced brain glucose uptake, whereas IR-deficient astrocytes show disturbed mitochondrial responses to glucose. While exploring the functional impact of disturbed mitochondrial function in astrocytes, we observed that GFAP-IR KO mice show uncoupling of brain blood flow with glucose uptake. Since IR-deficient astrocytes show higher levels of reactive oxidant species (ROS), this leads to stimulation of hypoxia-inducible factor-1α and, consequently, of the vascular endothelial growth factor angiogenic pathway. Indeed, GFAP-IR KO mice show disturbed brain vascularity and blood flow that is normalized by treatment with the antioxidant N-acetylcysteine (NAC). NAC ameliorated high ROS levels, normalized angiogenic signaling and mitochondrial function in IR-deficient astrocytes, and normalized neurovascular coupling in GFAP-IR KO mice. Our results indicate that by modulating glucose uptake and angiogenesis, insulin receptors in astrocytes participate in neurovascular coupling.


Subject(s)
Astrocytes , Brain , Insulin , Neovascularization, Physiologic , Neurovascular Coupling , Animals , Astrocytes/metabolism , Brain/blood supply , Glial Fibrillary Acidic Protein/genetics , Glucose/metabolism , Insulin/metabolism , Mice , Mice, Knockout , Reactive Oxygen Species/metabolism , Receptor, Insulin/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
7.
Trends Neurosci ; 44(10): 837-848, 2021 10.
Article in English | MEDLINE | ID: mdl-34334233

ABSTRACT

Activity-dependent long-term changes in synaptic transmission known as synaptic plasticity are fundamental processes in brain function and are recognized as the cellular basis of learning and memory. While the neuronal mechanisms underlying synaptic plasticity have been largely identified, the involvement of astrocytes in these processes has been less recognized. However, astrocytes are emerging as important cells that regulate synaptic function by interacting with neurons at tripartite synapses. In this review, we discuss recent evidence suggesting that astrocytes are necessary elements in long-term synaptic depression (LTD). We highlight the mechanistic heterogeneity of astrocyte contribution to this form of synaptic plasticity and propose that astrocytes are integral participants in LTD.


Subject(s)
Astrocytes , Depression , Humans , Neuronal Plasticity , Neurons , Synapses , Synaptic Transmission
8.
Neurobiol Dis ; 157: 105442, 2021 09.
Article in English | MEDLINE | ID: mdl-34246770

ABSTRACT

Neuregulin 1 (NRG1) and its receptor ERBB4 are schizophrenia (SZ) risk genes that control the development of both excitatory and inhibitory cortical circuits. Most studies focused on the characterization ErbB4 deficient mice. However, ErbB4 deletion concurrently perturbs the signaling of Nrg1 and Neuregulin 3 (Nrg3), another ligand expressed in the cortex. In addition, NRG1 polymorphisms linked to SZ locate mainly in non-coding regions and they may partially reduce Nrg1 expression. Here, to study the relevance of Nrg1 partial loss-of-function in cortical circuits we characterized a recently developed haploinsufficient mouse model of Nrg1 (Nrg1tm1Lex). These mice display SZ-like behavioral deficits. The cellular and molecular underpinnings of the behavioral deficits in Nrg1tm1Lex mice remain to be established. With multiple approaches including Magnetic Resonance Spectroscopy (MRS), electrophysiology, quantitative imaging and molecular analysis we found that Nrg1 haploinsufficiency impairs the inhibitory cortical circuits. We observed changes in the expression of molecules involved in GABAergic neurotransmission, decreased density of Vglut1 excitatory buttons onto Parvalbumin interneurons and decreased frequency of spontaneous inhibitory postsynaptic currents. Moreover, we found a decreased number of Parvalbumin positive interneurons in the cortex and altered expression of Calretinin. Interestingly, we failed to detect other alterations in excitatory neurons that were previously reported in ErbB4 null mice suggesting that the Nrg1 haploinsufficiency does not entirely phenocopies ErbB4 deletions. Altogether, this study suggests that Nrg1 haploinsufficiency primarily affects the cortical inhibitory circuits in the cortex and provides new insights into the structural and molecular synaptic impairment caused by NRG1 hypofunction in a preclinical model of SZ.


Subject(s)
Cerebral Cortex/metabolism , GABAergic Neurons/metabolism , Hippocampus/metabolism , Inhibitory Postsynaptic Potentials/genetics , Interneurons/metabolism , Neural Inhibition/genetics , Neuregulin-1/genetics , Vesicular Glutamate Transport Protein 1/metabolism , Animals , Calbindin 2/metabolism , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , GABAergic Neurons/pathology , Gene Expression , Haploinsufficiency , Hippocampus/diagnostic imaging , Hippocampus/pathology , Hippocampus/physiopathology , Interneurons/pathology , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Mice , Parvalbumins/metabolism , RNA, Messenger/metabolism , Receptor, ErbB-4/genetics , gamma-Aminobutyric Acid/metabolism
9.
Br J Pharmacol ; 178(17): 3395-3413, 2021 09.
Article in English | MEDLINE | ID: mdl-33830504

ABSTRACT

BACKGROUND AND PURPOSE: Activation of astrocytes contributes to synaptic remodelling, tissue repair and neuronal survival following traumatic brain injury (TBI). The mechanisms by which these cells interact to resident/infiltrated inflammatory cells to rewire neuronal networks and repair brain functions remain poorly understood. Here, we explored how TLR4-induced astrocyte activation modified synapses and cerebrovascular integrity following TBI. EXPERIMENTAL APPROACH: To determine how functional astrocyte alterations induced by activation of TLR4 pathway in inflammatory cells regulate synapses and neurovascular integrity after TBI, we used pharmacology, genetic approaches, live calcium imaging, immunofluorescence, flow cytometry, blood-brain barrier (BBB) integrity assessment and molecular and behavioural methods. KEY RESULTS: Shortly after a TBI, there is a recruitment of excitable and reactive astrocytes mediated by TLR4 pathway activation with detrimental effects on post-synaptic density-95 (PSD-95)/vesicular glutamate transporter 1 (VGLUT1) synaptic puncta, BBB integrity and neurological outcome. Pharmacological blockage of the TLR4 pathway with resatorvid (TAK-242) partially reversed many of the observed effects. Synapses and BBB recovery after resatorvid administration were not observed in IP3 R2-/- mice, indicating that effects of TLR4 inhibition depend on the subsequent astrocyte activation. In addition, TBI increased the astrocytic-protein thrombospondin-1 necessary to induce a synaptic recovery in a sub-acute phase. CONCLUSIONS AND IMPLICATIONS: Our data demonstrate that TLR4-mediated signalling, most probably through microglia and/or infiltrated monocyte-astrocyte communication, plays a crucial role in the TBI pathophysiology and that its inhibition prevents synaptic loss and BBB damage accelerating tissue recovery/repair, which might represent a therapeutic potential in CNS injuries and disorders.


Subject(s)
Astrocytes , Brain Injuries, Traumatic , Animals , Astrocytes/metabolism , Blood-Brain Barrier/metabolism , Mice , Microglia/metabolism , Neurons/metabolism , Toll-Like Receptor 4/metabolism
10.
Nat Commun ; 11(1): 782, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32034128

ABSTRACT

Food addiction is linked to obesity and eating disorders and is characterized by a loss of behavioral control and compulsive food intake. Here, using a food addiction mouse model, we report that the lack of cannabinoid type-1 receptor in dorsal telencephalic glutamatergic neurons prevents the development of food addiction-like behavior, which is associated with enhanced synaptic excitatory transmission in the medial prefrontal cortex (mPFC) and in the nucleus accumbens (NAc). In contrast, chemogenetic inhibition of neuronal activity in the mPFC-NAc pathway induces compulsive food seeking. Transcriptomic analysis and genetic manipulation identified that increased dopamine D2 receptor expression in the mPFC-NAc pathway promotes the addiction-like phenotype. Our study unravels a new neurobiological mechanism underlying resilience and vulnerability to the development of food addiction, which could pave the way towards novel and efficient interventions for this disorder.


Subject(s)
Food Addiction/physiopathology , Nucleus Accumbens/physiology , Prefrontal Cortex/physiology , Receptors, Dopamine D2/genetics , Animals , Disease Models, Animal , Feeding Behavior/physiology , Food Addiction/genetics , Gene Expression Profiling , Gene Expression Regulation , Mice, Knockout , Neural Pathways/physiology , Receptor, Cannabinoid, CB1/genetics , Synaptic Transmission , Up-Regulation
12.
Cereb Cortex ; 30(2): 505-524, 2020 03 21.
Article in English | MEDLINE | ID: mdl-31240311

ABSTRACT

Phosphatase and tensin homolog on chromosome 10 (PTEN) is a tumor suppressor and autism-associated gene that exerts an important influence over neuronal structure and function during development. In addition, it participates in synaptic plasticity processes in adulthood. As an attempt to assess synaptic and developmental mechanisms by which PTEN can modulate cognitive function, we studied the consequences of 2 different genetic manipulations in mice: presence of additional genomic copies of the Pten gene (Ptentg) and knock-in of a truncated Pten gene lacking its PDZ motif (Pten-ΔPDZ), which is required for interaction with synaptic proteins. Ptentg mice exhibit substantial microcephaly, structural hypoconnectivity, enhanced synaptic depression at cortico-amygdala synapses, reduced anxiety, and intensified social interactions. In contrast, Pten-ΔPDZ mice have a much more restricted phenotype, with normal synaptic connectivity, but impaired synaptic depression at cortico-amygdala synapses and virtually abolished social interactions. These results suggest that synaptic actions of PTEN in the amygdala contribute to specific behavioral traits, such as sociability. Also, PTEN appears to function as a bidirectional rheostat in the amygdala: reduction in PTEN activity at synapses is associated with less sociability, whereas enhanced PTEN activity accompanies hypersocial behavior.


Subject(s)
Amygdala/physiology , Cerebral Cortex/physiology , Neuronal Plasticity , PTEN Phosphohydrolase/physiology , Social Behavior , Amygdala/ultrastructure , Animals , Female , Hippocampus/physiology , Male , Memory/physiology , Mice, Transgenic , Synapses/physiology , Synapses/ultrastructure
13.
Nat Commun ; 10(1): 2968, 2019 07 04.
Article in English | MEDLINE | ID: mdl-31273206

ABSTRACT

NMDA receptor-dependent long-term depression (LTD) in the hippocampus is a well-known form of synaptic plasticity that has been linked to different cognitive functions. The core mechanism for this form of plasticity is thought to be entirely neuronal. However, we now demonstrate that astrocytic activity drives LTD at CA3-CA1 synapses. We have found that LTD induction enhances astrocyte-to-neuron communication mediated by glutamate, and that Ca2+ signaling and SNARE-dependent vesicular release from the astrocyte are required for LTD expression. In addition, using optogenetic techniques, we show that low-frequency astrocytic activation, in the absence of presynaptic activity, is sufficient to induce postsynaptic AMPA receptor removal and LTD expression. Using cell-type-specific gene deletion, we show that astrocytic p38α MAPK is required for the increased astrocytic glutamate release and astrocyte-to-neuron communication during low-frequency stimulation. Accordingly, removal of astrocytic (but not neuronal) p38α abolishes LTD expression. Finally, this mechanism modulates long-term memory in vivo.


Subject(s)
Astrocytes/enzymology , Hippocampus/physiology , Memory, Long-Term/physiology , Mitogen-Activated Protein Kinase 14/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Behavior, Animal/physiology , Conditioning, Psychological/physiology , Fear/physiology , Female , Glutamic Acid/metabolism , Hippocampus/cytology , Long-Term Synaptic Depression/physiology , Male , Mice , Mice, Inbred C57BL , Neurons/physiology , Optogenetics , Patch-Clamp Techniques , Synaptic Potentials/physiology
14.
Glia ; 67(5): 915-934, 2019 05.
Article in English | MEDLINE | ID: mdl-30632636

ABSTRACT

Optogenetics has been widely expanded to enhance or suppress neuronal activity and it has been recently applied to glial cells. Here, we have used a new approach based on selective expression of melanopsin, a G-protein-coupled photopigment, in astrocytes to trigger Ca2+ signaling. Using the genetically encoded Ca2+ indicator GCaMP6f and two-photon imaging, we show that melanopsin is both competent to stimulate robust IP3-dependent Ca2+ signals in astrocyte fine processes, and to evoke an ATP/Adenosine-dependent transient boost of hippocampal excitatory synaptic transmission. Additionally, under low-frequency light stimulation conditions, melanopsin-transfected astrocytes can trigger long-term synaptic changes. In vivo, melanopsin-astrocyte activation enhances episodic-like memory, suggesting melanopsin as an optical tool that could recapitulate the wide range of regulatory actions of astrocytes on neuronal networks in behaving animals. These results describe a novel approach using melanopsin as a precise trigger for astrocytes that mimics their endogenous G-protein signaling pathways, and present melanopsin as a valuable optical tool for neuron-glia studies.


Subject(s)
Astrocytes/metabolism , Nerve Net/metabolism , Neurons/metabolism , Optogenetics/methods , Rod Opsins/metabolism , 2-Amino-5-phosphonovalerate/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Alanine/analogs & derivatives , Alanine/pharmacology , Animals , Azo Compounds/pharmacology , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Chelating Agents/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/cytology , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Light , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pyridoxal Phosphate/analogs & derivatives , Pyridoxal Phosphate/pharmacology , Pyrimidines/pharmacology , Rod Opsins/genetics , Synaptic Potentials/physiology , Triazoles/pharmacology , Xanthenes/pharmacology
15.
J Vis Exp ; (120)2017 02 15.
Article in English | MEDLINE | ID: mdl-28287556

ABSTRACT

The nervous system is composed of an enormous range of distinct neuronal types. These neuronal subpopulations are characterized by, among other features, their distinct dendritic morphologies, their specific patterns of axonal connectivity, and their selective firing responses. The molecular and cellular mechanisms responsible for these aspects of differentiation during development are still poorly understood. Here, we describe combined protocols for labeling and characterizing the structural connectivity and excitability of cortical neurons. Modification of the in utero electroporation (IUE) protocol allows the labeling of a sparse population of neurons. This, in turn, enables the identification and tracking of the dendrites and axons of individual neurons, the precise characterization of the laminar location of axonal projections, and morphometric analysis. IUE can also be used to investigate changes in the excitability of wild-type (WT) or genetically modified neurons by combining it with whole-cell recording from acute slices of electroporated brains. These two techniques contribute to a better understanding of the coupling of structural and functional connectivity and of the molecular mechanisms controlling neuronal diversity during development. These developmental processes have important implications on axonal wiring, the functional diversity of neurons, and the biology of cognitive disorders.


Subject(s)
Cerebral Cortex/physiology , Electroporation/methods , Neurons/physiology , Animals , Axons/physiology , Cerebral Cortex/cytology , Dendrites/physiology , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Models, Animal , Patch-Clamp Techniques
16.
EMBO J ; 35(13): 1417-36, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27198172

ABSTRACT

Tau is a microtubule-associated neuronal protein found mainly in axons. However, its presence in dendrites and dendritic spines is particularly relevant due to its involvement in synaptic plasticity and neurodegeneration. Here, we show that Tau plays a novel in vivo role in the morphological and synaptic maturation of newborn hippocampal granule neurons under basal conditions. Furthermore, we reveal that Tau is involved in the selective cell death of immature granule neurons caused by acute stress. Also, Tau deficiency protects newborn neurons from the stress-induced dendritic atrophy and loss of postsynaptic densities (PSDs). Strikingly, we also demonstrate that Tau regulates the increase in newborn neuron survival triggered by environmental enrichment (EE). Moreover, newborn granule neurons from Tau(-/-) mice did not show any stimulatory effect of EE on dendritic development or on PSD generation. Thus, our data demonstrate that Tau(-/-) mice show impairments in the maturation of newborn granule neurons under basal conditions and that they are insensitive to the modulation of adult hippocampal neurogenesis exerted by both stimulatory and detrimental stimuli.


Subject(s)
Hippocampus/cytology , Hippocampus/physiology , Neurogenesis , tau Proteins/metabolism , Animals , Mice , Mice, Knockout
17.
Neuron ; 89(3): 494-506, 2016 Feb 03.
Article in English | MEDLINE | ID: mdl-26804994

ABSTRACT

Neuronal subtype-specific transcription factors (TFs) instruct key features of neuronal function and connectivity. Activity-dependent mechanisms also contribute to wiring and circuit assembly, but whether and how they relate to TF-directed neuronal differentiation is poorly investigated. Here we demonstrate that the TF Cux1 controls the formation of the layer II/III corpus callosum (CC) projections through the developmental transcriptional regulation of Kv1 voltage-dependent potassium channels and the resulting postnatal switch to a Kv1-dependent firing mode. Loss of Cux1 function led to a decrease in the expression of Kv1 transcripts, aberrant firing responses, and selective loss of CC contralateral innervation. Firing and innervation were rescued by re-expression of Kv1 or postnatal reactivation of Cux1. Knocking down Kv1 mimicked Cux1-mediated CC axonal loss. These findings reveal that activity-dependent processes are central bona fide components of neuronal TF-differentiation programs and establish the importance of intrinsic firing modes in circuit assembly within the neocortex.


Subject(s)
Action Potentials/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Neurons/physiology , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Shaker Superfamily of Potassium Channels/physiology , Animals , Corpus Callosum/cytology , Corpus Callosum/growth & development , Corpus Callosum/physiology , Gene Knockdown Techniques , Mice , Mice, Transgenic , Primary Cell Culture , Shaker Superfamily of Potassium Channels/biosynthesis , Shaker Superfamily of Potassium Channels/genetics
18.
Cereb Cortex ; 25(10): 3699-712, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25260706

ABSTRACT

Endocannabinoids (eCBs) play key roles in brain function, acting as modulatory signals in synaptic transmission and plasticity. They are recognized as retrograde messengers that mediate long-term synaptic depression (LTD), but their ability to induce long-term potentiation (LTP) is poorly known. We show that eCBs induce the long-term enhancement of transmitter release at single hippocampal synapses through stimulation of astrocytes when coincident with postsynaptic activity. This LTP requires the coordinated activity of the 3 elements of the tripartite synapse: 1) eCB-evoked astrocyte calcium signal that stimulates glutamate release; 2) postsynaptic nitric oxide production; and 3) activation of protein kinase C and presynaptic group I metabotropic glutamate receptors, whose location at presynaptic sites was confirmed by immunoelectron microscopy. Hence, while eCBs act as retrograde signals to depress homoneuronal synapses, they serve as lateral messengers to induce LTP in distant heteroneuronal synapses through stimulation of astrocytes. Therefore, eCBs can trigger LTP through stimulation of astrocyte-neuron signaling, revealing novel cellular mechanisms of eCB effects on synaptic plasticity.


Subject(s)
Astrocytes/physiology , Endocannabinoids/metabolism , Hippocampus/physiology , Long-Term Potentiation , Pyramidal Cells/physiology , Animals , Astrocytes/metabolism , Calcium/metabolism , Excitatory Postsynaptic Potentials , Hippocampus/metabolism , Mice, Inbred C57BL , Nitric Oxide/metabolism , Protein Kinase C/metabolism , Pyramidal Cells/metabolism , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction , Synapses/metabolism
19.
J Neurosci ; 34(38): 12738-44, 2014 Sep 17.
Article in English | MEDLINE | ID: mdl-25232111

ABSTRACT

Experience-dependent plasticity of synaptic transmission, which represents the cellular basis of learning, is accompanied by morphological changes in dendritic spines. Astrocytic processes are intimately associated with synapses, structurally enwrapping and functionally interacting with dendritic spines and synaptic terminals by responding to neurotransmitters and by releasing gliotransmitters that regulate synaptic function. While studies on structural synaptic plasticity have focused on neuronal elements, the structural-functional plasticity of astrocyte-neuron relationships remains poorly known. Here we show that stimuli inducing hippocampal synaptic LTP enhance the motility of synapse-associated astrocytic processes. This motility increase is relatively rapid, starting <5 min after the stimulus, and reaching a maximum in 20-30 min (t(1/2) = 10.7 min). It depends on presynaptic activity and requires G-protein-mediated Ca(2+) elevations in astrocytes. The structural remodeling is accompanied by changes in the ability of astrocytes to regulate synaptic transmission. Sensory stimuli that increase astrocyte Ca(2+) also induce similar plasticity in mouse somatosensory cortex in vivo. Therefore, structural relationships between astrocytic processes and dendritic spines undergo activity-dependent changes with metaplasticity consequences on synaptic regulation. These results reveal novel forms of synaptic plasticity based on structural-functional changes of astrocyte-neuron interactions.


Subject(s)
Astrocytes/physiology , Dendritic Spines/physiology , Neuronal Plasticity/physiology , Action Potentials/physiology , Animals , Astrocytes/cytology , Astrocytes/metabolism , Calcium/metabolism , Female , Hippocampus/physiology , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Mice , Somatosensory Cortex/physiology , Synaptic Transmission/physiology
20.
Philos Trans R Soc Lond B Biol Sci ; 369(1654): 20130599, 2014 Oct 19.
Article in English | MEDLINE | ID: mdl-25225093

ABSTRACT

Astrocytes are emerging as integral functional components of synapses, responding to synaptically released neurotransmitters and regulating synaptic transmission and plasticity. Thus, they functionally interact with neurons establishing tripartite synapses: a functional concept that refers to the existence of communication between astrocytes and neurons and its crucial role in synaptic function. Here, we discuss recent evidence showing that astrocytes are involved in the endocannabinoid (ECB) system, responding to exogenous cannabinoids as well as ECBs through activation of type 1 cannabinoid receptors, which increase intracellular calcium and stimulate the release of glutamate that modulates synaptic transmission and plasticity. We also discuss the consequences of ECB signalling in tripartite synapses on the astrocyte-mediated regulation of synaptic function, which reveal novel properties of synaptic regulation by ECBs, such as the spatially controlled dual effect on synaptic strength and the lateral potentiation of synaptic efficacy. Finally, we discuss the potential implications of ECB signalling for astrocytes in brain pathology and animal behaviour.


Subject(s)
Astrocytes/physiology , Endocannabinoids/metabolism , Models, Neurological , Neurotransmitter Agents/metabolism , Receptors, Cannabinoid/metabolism , Signal Transduction/physiology , Synapses/physiology , Astrocytes/metabolism , Humans , Neuronal Plasticity/physiology , Synapses/metabolism , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...