Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep ; 43(1): 113595, 2024 01 23.
Article in English | MEDLINE | ID: mdl-38117654

ABSTRACT

Cellular signaling involves a large repertoire of membrane receptors operating in overlapping spatiotemporal regimes and targeting many common intracellular effectors. However, both the molecular mechanisms and the physiological roles of crosstalk between receptors, especially those from different superfamilies, are poorly understood. We find that the receptor tyrosine kinase (RTK) TrkB and the G-protein-coupled receptor (GPCR) metabotropic glutamate receptor 5 (mGluR5) together mediate hippocampal synaptic plasticity in response to brain-derived neurotrophic factor (BDNF). Activated TrkB enhances constitutive mGluR5 activity to initiate a mode switch that drives BDNF-dependent sustained, oscillatory Ca2+ signaling and enhanced MAP kinase activation. This crosstalk is mediated, in part, by synergy between Gßγ, released by TrkB, and Gαq-GTP, released by mGluR5, to enable physiologically relevant RTK/GPCR crosstalk.


Subject(s)
Brain-Derived Neurotrophic Factor , Receptor Protein-Tyrosine Kinases , Signal Transduction/physiology , Receptor, trkB/metabolism , Receptors, G-Protein-Coupled , Neuronal Plasticity/physiology
2.
bioRxiv ; 2023 Aug 28.
Article in English | MEDLINE | ID: mdl-37693535

ABSTRACT

Cellular signaling involves a large repertoire of membrane receptors operating in overlapping spatiotemporal regimes and targeting many common intracellular effectors. However, both the molecular mechanisms and physiological roles of crosstalk between receptors, especially those from different superfamilies, are poorly understood. We find that the receptor tyrosine kinase (RTK), TrkB, and the G protein-coupled receptor (GPCR), metabotropic glutamate receptor 5 (mGluR5), together mediate a novel form of hippocampal synaptic plasticity in response to brain-derived neurotrophic factor (BDNF). Activated TrkB enhances constitutive mGluR5 activity to initiate a mode-switch that drives BDNF-dependent sustained, oscillatory Ca 2+ signaling and enhanced MAP kinase activation. This crosstalk is mediated, in part, by synergy between Gßγ, released by TrkB, and Gα q -GTP, released by mGluR5, to enable a previously unidentified form of physiologically relevant RTK/GPCR crosstalk.

3.
Proc Natl Acad Sci U S A ; 117(48): 30710-30721, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33208539

ABSTRACT

Although ubiquitous in biological studies, the enhanced green and yellow fluorescent proteins (EGFP and EYFP) were not specifically optimized for neuroscience, and their underwhelming brightness and slow expression in brain tissue limits the fidelity of dendritic spine analysis and other indispensable techniques for studying neurodevelopment and plasticity. We hypothesized that EGFP's low solubility in mammalian systems must limit the total fluorescence output of whole cells, and that improving folding efficiency could therefore translate into greater brightness of expressing neurons. By introducing rationally selected combinations of folding-enhancing mutations into GFP templates and screening for brightness and expression rate in human cells, we developed mGreenLantern, a fluorescent protein having up to sixfold greater brightness in cells than EGFP. mGreenLantern illuminates neurons in the mouse brain within 72 h, dramatically reducing lag time between viral transduction and imaging, while its high brightness improves detection of neuronal morphology using widefield, confocal, and two-photon microscopy. When virally expressed to projection neurons in vivo, mGreenLantern fluorescence developed four times faster than EYFP and highlighted long-range processes that were poorly detectable in EYFP-labeled cells. Additionally, mGreenLantern retains strong fluorescence after tissue clearing and expansion microscopy, thereby facilitating superresolution and whole-brain imaging without immunohistochemistry. mGreenLantern can directly replace EGFP/EYFP in diverse systems due to its compatibility with GFP filter sets, recognition by EGFP antibodies, and excellent performance in mouse, human, and bacterial cells. Our screening and rational engineering approach is broadly applicable and suggests that greater potential of fluorescent proteins, including biosensors, could be unlocked using a similar strategy.


Subject(s)
Gene Expression , Green Fluorescent Proteins/genetics , Molecular Imaging , Neurons/metabolism , Animals , Brain/metabolism , Fluorescent Antibody Technique , Genes, Reporter , Green Fluorescent Proteins/chemistry , Mice , Microscopy, Fluorescence , Molecular Imaging/methods , Mutation , Protein Stability , Proteolysis , Solubility , Spectrum Analysis
4.
Addict Biol ; 22(6): 1706-1718, 2017 Nov.
Article in English | MEDLINE | ID: mdl-27457910

ABSTRACT

Caffeine has cognitive-enhancing properties with effects on learning and memory, concentration, arousal and mood. These effects imply changes at circuital and synaptic level, but the mechanism by which caffeine modifies synaptic plasticity remains elusive. Here we report that caffeine, at concentrations representing moderate to high levels of consumption in humans, induces an NMDA receptor-independent form of LTP (CAF LTP) in the CA1 region of the hippocampus by promoting calcium-dependent secretion of BDNF, which subsequently activates TrkB-mediated signaling required for the expression of CAF LTP. Our data include the novel observation that insulin receptor substrate 2 (IRS2) is phosphorylated during induction of CAF LTP, a process that requires cytosolic free Ca2+ . Consistent with the involvement of IRS2 signals in caffeine-mediated synaptic plasticity, phosphorylation of Akt (Ser473) in response to LTP induction is defective in Irs2-/- mice, demonstrating that these plasticity changes are associated with downstream targets of the phosphoinositide 3-kinase (PI3K) pathway. These findings indicate that TrkB-IRS2 signals are essential for activation of PI3K during the induction of LTP by caffeine.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Caffeine/pharmacology , Central Nervous System Stimulants/pharmacology , Insulin Receptor Substrate Proteins/metabolism , Neuronal Plasticity/drug effects , Signal Transduction/drug effects , Animals , Brain-Derived Neurotrophic Factor/drug effects , Female , Insulin Receptor Substrate Proteins/drug effects , Insulin Receptor Substrate Proteins/genetics , Male , Mice , Models, Animal
5.
Nat Commun ; 6: 8518, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26439639

ABSTRACT

Myelin regeneration can occur spontaneously in demyelinating diseases such as multiple sclerosis (MS). However, the underlying mechanisms and causes of its frequent failure remain incompletely understood. Here we show, using an in-vivo remyelination model, that demyelinated axons are electrically active and generate de novo synapses with recruited oligodendrocyte progenitor cells (OPCs), which, early after lesion induction, sense neuronal activity by expressing AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid)/kainate receptors. Blocking neuronal activity, axonal vesicular release or AMPA receptors in demyelinated lesions results in reduced remyelination. In the absence of neuronal activity there is a ∼6-fold increase in OPC number within the lesions and a reduced proportion of differentiated oligodendrocytes. These findings reveal that neuronal activity and release of glutamate instruct OPCs to differentiate into new myelinating oligodendrocytes that recover lost function. Co-localization of OPCs with the presynaptic protein VGluT2 in MS lesions implies that this mechanism may provide novel targets to therapeutically enhance remyelination.


Subject(s)
Axons/metabolism , Glutamic Acid/metabolism , Multiple Sclerosis/metabolism , Myelin Sheath/metabolism , Neurons/metabolism , Oligodendroglia/metabolism , Receptors, AMPA/metabolism , Regeneration/physiology , Action Potentials , Adult , Animals , Brain/metabolism , Female , Humans , Immunohistochemistry , Male , Microscopy, Electron , Middle Aged , Multiple Sclerosis/pathology , Myelin Sheath/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, Kainic Acid/metabolism , Stem Cells , Vesicular Glutamate Transport Protein 2/metabolism
6.
Neurobiol Dis ; 52: 117-27, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23220201

ABSTRACT

The cognitive dysfunctions of Down Syndrome (DS) individuals are the most disabling alterations caused by the trisomy of human chromosome 21 (HSA21). In trisomic Ts65Dn mice, a genetic model for DS, the overexpression of HSA21 homologous genes has been associated with strong visuo-spatial cognitive alterations, ascribed to hippocampal dysfunction. In the present study, we evaluated whether the normalization of the expression levels of Dyrk1A (Dual specificity tyrosine-phosphorylation-regulated kinase 1A), a candidate gene for DS, might correct hippocampal defects in Ts65Dn mice. In the hippocampus of 2 month-old Ts65Dn mice, such normalization was achieved through the stereotaxical injection of adeno-associated viruses containing a short hairpin RNA against Dyrk1A (AAV2/1-shDyrk1A) and a luciferase reporter gene. The injected hippocampi were efficiently transduced, as shown by bioluminescence in vivo imaging, luciferase activity quantification and immunohistochemical analysis. At the molecular level, viral infusion allowed the normalization of the targeted Dyrk1A expression, as well as of the key players of the MAPK/CREB pathway. The electrophysiological recordings of hippocampal slices from Ts65Dn mice injected with AAV2/1-shDyrk1A displayed attenuation of the synaptic plasticity defects of trisomic mice. In contrast, contralateral hippocampal injection with an AAV2/1 control virus containing a scrambled sequence, showed neither the normalization of Dyrk1A levels nor changes of synaptic plasticity. In the Morris water maze task, although long-term consolidation of the task was not achieved, treated Ts65Dn mice displayed initially a normalized thigmotactic behavior, similar to euploid littermates, indicating the partial improvement in their hippocampal-dependent search strategy. Taken together, these results show Dyrk1A as a critical player in the pathophysiology of DS and define Dyrk1A as a therapeutic target in adult trisomic mice.


Subject(s)
Down Syndrome/physiopathology , Hippocampus/physiopathology , Maze Learning/physiology , Neuronal Plasticity/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Synapses/genetics , Animals , Behavior, Animal/physiology , Dependovirus , Disease Models, Animal , Down Syndrome/genetics , Down Syndrome/metabolism , Hippocampus/metabolism , Mice , Mice, Transgenic , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Dyrk Kinases
7.
Mol Neurodegener ; 6(1): 71, 2011 Oct 10.
Article in English | MEDLINE | ID: mdl-21985529

ABSTRACT

BACKGROUND: Brain-Derived Neurotrophic Factor (BDNF) is the main candidate for neuroprotective therapy for Huntington's disease (HD), but its conditional administration is one of its most challenging problems. RESULTS: Here we used transgenic mice that over-express BDNF under the control of the Glial Fibrillary Acidic Protein (GFAP) promoter (pGFAP-BDNF mice) to test whether up-regulation and release of BDNF, dependent on astrogliosis, could be protective in HD. Thus, we cross-mated pGFAP-BDNF mice with R6/2 mice to generate a double-mutant mouse with mutant huntingtin protein and with a conditional over-expression of BDNF, only under pathological conditions. In these R6/2:pGFAP-BDNF animals, the decrease in striatal BDNF levels induced by mutant huntingtin was prevented in comparison to R6/2 animals at 12 weeks of age. The recovery of the neurotrophin levels in R6/2:pGFAP-BDNF mice correlated with an improvement in several motor coordination tasks and with a significant delay in anxiety and clasping alterations. Therefore, we next examined a possible improvement in cortico-striatal connectivity in R62:pGFAP-BDNF mice. Interestingly, we found that the over-expression of BDNF prevented the decrease of cortico-striatal presynaptic (VGLUT1) and postsynaptic (PSD-95) markers in the R6/2:pGFAP-BDNF striatum. Electrophysiological studies also showed that basal synaptic transmission and synaptic fatigue both improved in R6/2:pGAP-BDNF mice. CONCLUSIONS: These results indicate that the conditional administration of BDNF under the GFAP promoter could become a therapeutic strategy for HD due to its positive effects on synaptic plasticity.

8.
Epilepsia ; 51(3): 384-90, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19732136

ABSTRACT

PURPOSE: In this study, we explore the antiepileptic effects of flufenamic acid (FFA) in order to identify the cellular mechanisms that underlie the potential anticonvulsant properties of this nonsteroidal antiinflammatory compound. METHODS: The mechanisms of FFA action were analyzed using an in vitro model in which epileptiform activity was induced in hippocampal slices by perfusion with 100 microm 4-aminopyridine (4-AP) added to a modified Mg(2+)-free solution. The activity of CA1 pyramidal neurons as well as the synaptic connection between CA3 and CA1 was monitored using extracellular and patch-clamp recordings. RESULTS: Epileptiform activity was suppressed in hippocampal neurons by FFA at concentrations between 50 and 200 microm. Glutamatergic excitatory synaptic transmission was diminished by FFA without modifying recurrent gamma-aminobutyric acid (GABA)ergic synaptic inhibition. Several lines of evidence indicated that FFA did not decrease neurotransmitter release probability, implicating a postsynaptic mechanism of action. FFA also potently reduced neuronal excitability, but did not alter the amplitude, duration, or undershoot of action potentials. CONCLUSIONS: Our results suggest that FFA exerts an anticonvulsive effect on hippocampal pyramidal neurons by simultaneously decreasing glutamatergic excitatory synaptic activity and reducing neuronal excitability. Therefore, our study provides experimental evidence that FFA may represent an effective pharmacologic agent in the treatment of epilepsy in the mammalian central nervous system.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Anticonvulsants/pharmacology , Epilepsy/prevention & control , Epilepsy/physiopathology , Excitatory Postsynaptic Potentials/drug effects , Flufenamic Acid/pharmacology , Hippocampus/drug effects , Neurons/drug effects , Synaptic Transmission/drug effects , 4-Aminopyridine/pharmacology , Action Potentials/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Epilepsy/drug therapy , Hippocampus/physiology , Inhibitory Postsynaptic Potentials/drug effects , Neural Inhibition/drug effects , Neurons/physiology , Patch-Clamp Techniques , Pyramidal Cells/drug effects , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/drug effects , gamma-Aminobutyric Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...