Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Adv ; 9(31): eadg0666, 2023 08 02.
Article in English | MEDLINE | ID: mdl-37531435

ABSTRACT

The insulin superfamily of peptides is essential for homeostasis as well as neuronal plasticity, learning, and memory. Here, we show that insulin-like growth factors 1 and 2 (IGF1 and IGF2) are differentially expressed in hippocampal neurons and released in an activity-dependent manner. Using a new fluorescence resonance energy transfer sensor for IGF1 receptor (IGF1R) with two-photon fluorescence lifetime imaging, we find that the release of IGF1 triggers rapid local autocrine IGF1R activation on the same spine and more than several micrometers along the stimulated dendrite, regulating the plasticity of the activated spine in CA1 pyramidal neurons. In CA3 neurons, IGF2, instead of IGF1, is responsible for IGF1R autocrine activation and synaptic plasticity. Thus, our study demonstrates the cell type-specific roles of IGF1 and IGF2 in hippocampal plasticity and a plasticity mechanism mediated by the synthesis and autocrine signaling of IGF peptides in pyramidal neurons.


Subject(s)
Autocrine Communication , Dendritic Spines , Dendritic Spines/physiology , Hippocampus/physiology , Neuronal Plasticity/physiology , Pyramidal Cells/metabolism
2.
J Neurosci ; 43(30): 5432-5447, 2023 07 26.
Article in English | MEDLINE | ID: mdl-37277178

ABSTRACT

The activity-dependent plasticity of synapses is believed to be the cellular basis of learning. These synaptic changes are mediated through the coordination of local biochemical reactions in synapses and changes in gene transcription in the nucleus to modulate neuronal circuits and behavior. The protein kinase C (PKC) family of isozymes has long been established as critical for synaptic plasticity. However, because of a lack of suitable isozyme-specific tools, the role of the novel subfamily of PKC isozymes is largely unknown. Here, through the development of fluorescence lifetime imaging-fluorescence resonance energy transfer activity sensors, we investigate novel PKC isozymes in synaptic plasticity in CA1 pyramidal neurons of mice of either sex. We find that PKCδ is activated downstream of TrkB and DAG production, and that the spatiotemporal nature of its activation depends on the plasticity stimulation. In response to single-spine plasticity, PKCδ is activated primarily in the stimulated spine and is required for local expression of plasticity. However, in response to multispine stimulation, a long-lasting and spreading activation of PKCδ scales with the number of spines stimulated and, by regulating cAMP response-element binding protein activity, couples spine plasticity to transcription in the nucleus. Thus, PKCδ plays a dual functional role in facilitating synaptic plasticity.SIGNIFICANCE STATEMENT Synaptic plasticity, or the ability to change the strength of the connections between neurons, underlies learning and memory and is critical for brain health. The protein kinase C (PKC) family is central to this process. However, understanding how these kinases work to mediate plasticity has been limited by a lack of tools to visualize and perturb their activity. Here, we introduce and use new tools to reveal a dual role for PKCδ in facilitating local synaptic plasticity and stabilizing this plasticity through spine-to-nucleus signaling to regulate transcription. This work provides new tools to overcome limitations in studying isozyme-specific PKC function and provides insight into molecular mechanisms of synaptic plasticity.


Subject(s)
Isoenzymes , Signal Transduction , Animals , Mice , Signal Transduction/physiology , Synapses/physiology , Neuronal Plasticity/physiology , Protein Kinase C/metabolism
3.
eNeuro ; 8(1)2021.
Article in English | MEDLINE | ID: mdl-33139322

ABSTRACT

ADAP1/Centaurin-α1 (CentA1) functions as an Arf6 GTPase-activating protein highly enriched in the brain. Previous studies demonstrated the involvement of CentA1 in brain function as a regulator of dendritic differentiation and a potential mediator of Alzheimer's disease (AD) pathogenesis. To better understand the neurobiological functions of CentA1 signaling in the brain, we developed Centa1 knock-out (KO) mice. The KO animals showed neither brain development nor synaptic ultrastructure deficits in the hippocampus. However, they exhibited significantly higher density and enhanced structural plasticity of dendritic spines in the CA1 region of the hippocampus compared with non-transgenic (NTG) littermates. Moreover, the deletion of Centa1 improved performance in the object-in-place (OIP) spatial memory task. These results suggest that CentA1 functions as a negative regulator of spine density and plasticity, and of hippocampus-dependent memory formation. Thus, CentA1 and its downstream signaling may serve as a potential therapeutic target to prevent memory decline associated with aging and brain disorders.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Dendritic Spines , Hippocampus , Memory , Nerve Tissue Proteins/genetics , Alzheimer Disease , Animals , Dendritic Spines/metabolism , GTPase-Activating Proteins/metabolism , Hippocampus/metabolism , Mice
4.
Neuron ; 105(5): 799-812.e5, 2020 03 04.
Article in English | MEDLINE | ID: mdl-31883788

ABSTRACT

Sensory experiences cause long-term modifications of neuronal circuits by modulating activity-dependent transcription programs that are vital for regulation of long-term synaptic plasticity and memory. However, it has not been possible to precisely determine the interaction between neuronal activity patterns and transcription factor activity. Here we present a technique using two-photon fluorescence lifetime imaging (2pFLIM) with new FRET biosensors to chronically image in vivo signaling of CREB, an activity-dependent transcription factor important for synaptic plasticity, at single-cell resolution. Simultaneous imaging of the red-shifted CREB sensor and GCaMP permitted exploration of how experience shapes the interplay between CREB and neuronal activity in the neocortex of awake mice. Dark rearing increased the sensitivity of CREB activity to Ca2+ elevations and prolonged the duration of CREB activation to more than 24 h in the visual cortex. This technique will allow researchers to unravel the transcriptional dynamics underlying experience-dependent plasticity in the brain.


Subject(s)
Calcium/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Neocortex/metabolism , Neuronal Plasticity , Neurons/metabolism , Animals , Darkness , Fluorescence Resonance Energy Transfer , Mice , Neocortex/cytology , Neural Pathways , Neurons/cytology , Photic Stimulation , Signal Transduction , Single-Cell Analysis , Somatosensory Cortex/cytology , Somatosensory Cortex/metabolism , Visual Cortex/cytology , Visual Cortex/metabolism
5.
Nat Neurosci ; 21(8): 1027-1037, 2018 08.
Article in English | MEDLINE | ID: mdl-30013171

ABSTRACT

The protein kinase C (PKC) enzymes have long been established as critical for synaptic plasticity. However, it is unknown whether Ca2+-dependent PKC isozymes are activated in dendritic spines during plasticity and, if so, how this synaptic activity is encoded by PKC. Here, using newly developed, isozyme-specific sensors, we demonstrate that classical isozymes are activated to varying degrees and with distinct kinetics. PKCα is activated robustly and rapidly in stimulated spines and is the only isozyme required for structural plasticity. This specificity depends on a PDZ-binding motif present only in PKCα. The activation of PKCα during plasticity requires both NMDA receptor Ca2+ flux and autocrine brain-derived neurotrophic factor (BDNF)-TrkB signaling, two pathways that differ vastly in their spatiotemporal scales of signaling. Our results suggest that, by integrating these signals, PKCα combines a measure of recent, nearby synaptic plasticity with local synaptic input, enabling complex cellular computations such as heterosynaptic facilitation of plasticity necessary for efficient hippocampus-dependent learning.


Subject(s)
Autocrine Communication/physiology , Brain-Derived Neurotrophic Factor/physiology , Calcium Signaling/physiology , Neuronal Plasticity/physiology , Protein Kinase C-alpha/physiology , Animals , Autocrine Communication/genetics , Brain-Derived Neurotrophic Factor/genetics , Calcium Signaling/genetics , Dendritic Spines , Enzyme Activation , Hippocampus/physiology , Isoenzymes , Kinetics , Learning/physiology , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Kinase C-alpha/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
6.
eNeuro ; 5(3)2018.
Article in English | MEDLINE | ID: mdl-29911178

ABSTRACT

Pyramidal neurons in hippocampal area CA2 are distinct from neighboring CA1 in that they resist synaptic long-term potentiation (LTP) at CA3 Schaffer collateral synapses. Regulator of G protein signaling 14 (RGS14) is a complex scaffolding protein enriched in CA2 dendritic spines that naturally blocks CA2 synaptic plasticity and hippocampus-dependent learning, but the cellular mechanisms by which RGS14 gates LTP are largely unexplored. A previous study has attributed the lack of plasticity to higher rates of calcium (Ca2+) buffering and extrusion in CA2 spines. Additionally, a recent proteomics study revealed that RGS14 interacts with two key Ca2+-activated proteins in CA2 neurons: calcium/calmodulin and CaMKII. Here, we investigated whether RGS14 regulates Ca2+ signaling in its host CA2 neurons. We found that the nascent LTP of CA2 synapses caused by genetic knockout (KO) of RGS14 in mice requires Ca2+-dependent postsynaptic signaling through NMDA receptors, CaMK, and PKA, revealing similar mechanisms to those in CA1. We report that RGS14 negatively regulates the long-term structural plasticity of dendritic spines of CA2 neurons. We further show that wild-type (WT) CA2 neurons display significantly attenuated spine Ca2+ transients during structural plasticity induction compared with the Ca2+ transients from CA2 spines of RGS14 KO mice and CA1 controls. Finally, we demonstrate that acute overexpression of RGS14 is sufficient to block spine plasticity, and elevating extracellular Ca2+ levels restores plasticity to RGS14-expressing neurons. Together, these results demonstrate for the first time that RGS14 regulates plasticity in hippocampal area CA2 by restricting Ca2+ elevations in CA2 spines and downstream signaling pathways.


Subject(s)
CA2 Region, Hippocampal/physiology , Calcium Signaling , Long-Term Potentiation , Pyramidal Cells/physiology , RGS Proteins/physiology , Synapses/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Dendritic Spines/physiology , Female , Male , Mice, Knockout , Receptors, N-Methyl-D-Aspartate
8.
Neuron ; 94(4): 800-808.e4, 2017 May 17.
Article in English | MEDLINE | ID: mdl-28521133

ABSTRACT

CaMKII plays a critical role in decoding calcium (Ca2+) signals to initiate long-lasting synaptic plasticity. However, the properties of CaMKII that mediate Ca2+ signals in spines remain elusive. Here, we measured CaMKII activity in spines using fast-framing two-photon fluorescence lifetime imaging. Following each pulse during repetitive Ca2+ elevations, CaMKII activity increased in a stepwise manner. Thr286 phosphorylation slows the decay of CaMKII and thus lowers the frequency required to induce spine plasticity by several fold. In the absence of Thr286 phosphorylation, increasing the stimulation frequency results in high peak mutant CaMKIIT286A activity that is sufficient for inducing plasticity. Our findings demonstrate that Thr286 phosphorylation plays an important role in induction of LTP by integrating Ca2+ signals, and it greatly promotes, but is dispensable for, the activation of CaMKII and LTP.


Subject(s)
CA1 Region, Hippocampal/metabolism , Calcium Signaling/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium/metabolism , Long-Term Potentiation/physiology , Pyramidal Cells/metabolism , Animals , CA1 Region, Hippocampal/physiology , Hippocampus/metabolism , Hippocampus/physiology , Mice , Microscopy, Fluorescence , Neuronal Plasticity , Patch-Clamp Techniques , Phosphorylation , Pyramidal Cells/physiology
9.
Neuron ; 94(1): 37-47.e5, 2017 Apr 05.
Article in English | MEDLINE | ID: mdl-28318784

ABSTRACT

Elucidating temporal windows of signaling activity required for synaptic and behavioral plasticity is crucial for understanding molecular mechanisms underlying these phenomena. Here, we developed photoactivatable autocamtide inhibitory peptide 2 (paAIP2), a genetically encoded, light-inducible inhibitor of CaMKII activity. The photoactivation of paAIP2 in neurons for 1-2 min during the induction of LTP and structural LTP (sLTP) of dendritic spines inhibited these forms of plasticity in hippocampal slices of rodents. However, photoactivation ∼1 min after the induction did not affect them, suggesting that the initial 1 min of CaMKII activation is sufficient for inducing LTP and sLTP. Furthermore, the photoactivation of paAIP2 expressed in amygdalar neurons of mice during an inhibitory avoidance task revealed that CaMKII activity during, but not after, training is required for the memory formation. Thus, we demonstrated that paAIP2 is useful to elucidate the temporal window of CaMKII activation required for synaptic plasticity and learning.


Subject(s)
Avoidance Learning/physiology , CA1 Region, Hippocampal/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dendritic Spines/metabolism , Neuronal Plasticity/physiology , Pyramidal Cells/metabolism , Animals , Animals, Newborn , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Dendritic Spines/physiology , Electrophoresis, Polyacrylamide Gel , HEK293 Cells , HeLa Cells , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/physiology , Humans , Immunoblotting , Immunohistochemistry , Kinetics , Long-Term Potentiation/physiology , Mice , Microscopy, Fluorescence , Neurons/metabolism , Neurons/physiology , Optogenetics , Pyramidal Cells/physiology , RNA-Binding Proteins , Rats , Recombinant Fusion Proteins/genetics , Repressor Proteins , Tumor Suppressor Proteins/genetics
10.
Nature ; 538(7623): 99-103, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27680698

ABSTRACT

Brain-derived neurotrophic factor (BDNF) and its receptor TrkB are crucial for many forms of neuronal plasticity, including structural long-term potentiation (sLTP), which is a correlate of an animal's learning. However, it is unknown whether BDNF release and TrkB activation occur during sLTP, and if so, when and where. Here, using a fluorescence resonance energy transfer-based sensor for TrkB and two-photon fluorescence lifetime imaging microscopy, we monitor TrkB activity in single dendritic spines of CA1 pyramidal neurons in cultured murine hippocampal slices. In response to sLTP induction, we find fast (onset < 1 min) and sustained (>20 min) activation of TrkB in the stimulated spine that depends on NMDAR (N-methyl-d-aspartate receptor) and CaMKII signalling and on postsynaptically synthesized BDNF. We confirm the presence of postsynaptic BDNF using electron microscopy to localize endogenous BDNF to dendrites and spines of hippocampal CA1 pyramidal neurons. Consistent with these findings, we also show rapid, glutamate-uncaging-evoked, time-locked BDNF release from single dendritic spines using BDNF fused to superecliptic pHluorin. We demonstrate that this postsynaptic BDNF-TrkB signalling pathway is necessary for both structural and functional LTP. Together, these findings reveal a spine-autonomous, autocrine signalling mechanism involving NMDAR-CaMKII-dependent BDNF release from stimulated dendritic spines and subsequent TrkB activation on these same spines that is crucial for structural and functional plasticity.


Subject(s)
Autocrine Communication , Brain-Derived Neurotrophic Factor/metabolism , Dendritic Spines/metabolism , Membrane Glycoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dendritic Spines/ultrastructure , Enzyme Activation , Female , Fluorescence Resonance Energy Transfer , Glutamic Acid/metabolism , Green Fluorescent Proteins , HeLa Cells , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/ultrastructure , Humans , Long-Term Potentiation , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron , Microscopy, Fluorescence, Multiphoton , Post-Synaptic Density/metabolism , Pyramidal Cells/metabolism , Pyramidal Cells/ultrastructure , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Tissue Culture Techniques
11.
Science ; 342(6162): 1107-11, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-24288335

ABSTRACT

The late phase of long-term potentiation (LTP) at glutamatergic synapses, which is thought to underlie long-lasting memory, requires gene transcription in the nucleus. However, the mechanism by which signaling initiated at synapses is transmitted into the nucleus to induce transcription has remained elusive. Here, we found that induction of LTP in only three to seven dendritic spines in rat CA1 pyramidal neurons was sufficient to activate extracellular signal-regulated kinase (ERK) in the nucleus and regulate downstream transcription factors. Signaling from individual spines was integrated over a wide range of time (>30 minutes) and space (>80 micrometers). Spatially dispersed inputs over multiple branches activated nuclear ERK much more efficiently than clustered inputs over one branch. Thus, biochemical signals from individual dendritic spines exert profound effects on nuclear signaling.


Subject(s)
CA1 Region, Hippocampal/physiology , Dendritic Spines/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Long-Term Potentiation , Animals , CA1 Region, Hippocampal/enzymology , Cells, Cultured , Dendritic Spines/enzymology , Glutamates/metabolism , Rats , Signal Transduction , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...