Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Proc Natl Acad Sci U S A ; 120(37): e2308891120, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37669379

ABSTRACT

SYNGAP1 is a Ras-GTPase-activating protein highly enriched at excitatory synapses in the brain. De novo loss-of-function mutations in SYNGAP1 are a major cause of genetically defined neurodevelopmental disorders (NDDs). These mutations are highly penetrant and cause SYNGAP1-related intellectual disability (SRID), an NDD characterized by cognitive impairment, social deficits, early-onset seizures, and sleep disturbances. Studies in rodent neurons have shown that Syngap1 regulates developing excitatory synapse structure and function, and heterozygous Syngap1 knockout mice have deficits in synaptic plasticity, learning, and memory and have seizures. However, how specific SYNGAP1 mutations found in humans lead to disease has not been investigated in vivo. To explore this, we utilized the CRISPR-Cas9 system to generate knock-in mouse models with two distinct known causal variants of SRID: one with a frameshift mutation leading to a premature stop codon, SYNGAP1; L813RfsX22, and a second with a single-nucleotide mutation in an intron that creates a cryptic splice acceptor site leading to premature stop codon, SYNGAP1; c.3583-9G>A. While reduction in Syngap1 mRNA varies from 30 to 50% depending on the specific mutation, both models show ~50% reduction in Syngap1 protein, have deficits in synaptic plasticity, and recapitulate key features of SRID including hyperactivity and impaired working memory. These data suggest that half the amount of SYNGAP1 protein is key to the pathogenesis of SRID. These results provide a resource to study SRID and establish a framework for the development of therapeutic strategies for this disorder.


Subject(s)
Epilepsy , Intellectual Disability , Humans , Animals , Mice , Codon, Nonsense , Seizures , Brain , Disease Models, Animal , Memory Disorders , ras GTPase-Activating Proteins
2.
J Neurosci ; 44(11)2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38316559

ABSTRACT

Transcranial focused ultrasound stimulation (tFUS) is a noninvasive neuromodulation technique, which can penetrate deeper and modulate neural activity with a greater spatial resolution (on the order of millimeters) than currently available noninvasive brain stimulation methods, such as transcranial magnetic stimulation (TMS) and transcranial direct current stimulation (tDCS). While there are several studies demonstrating the ability of tFUS to modulate neuronal activity, it is unclear whether it can be used for producing long-term plasticity as needed to modify circuit function, especially in adult brain circuits with limited plasticity such as the thalamocortical synapses. Here we demonstrate that transcranial low-intensity focused ultrasound (LIFU) stimulation of the visual thalamus (dorsal lateral geniculate nucleus, dLGN), a deep brain structure, leads to NMDA receptor (NMDAR)-dependent long-term depression of its synaptic transmission onto layer 4 neurons in the primary visual cortex (V1) of adult mice of both sexes. This change is not accompanied by large increases in neuronal activity, as visualized using the cFos Targeted Recombination in Active Populations (cFosTRAP2) mouse line, or activation of microglia, which was assessed with IBA-1 staining. Using a model (SONIC) based on the neuronal intramembrane cavitation excitation (NICE) theory of ultrasound neuromodulation, we find that the predicted activity pattern of dLGN neurons upon sonication is state-dependent with a range of activity that falls within the parameter space conducive for inducing long-term synaptic depression. Our results suggest that noninvasive transcranial LIFU stimulation has a potential for recovering long-term plasticity of thalamocortical synapses in the postcritical period adult brain.


Subject(s)
Transcranial Direct Current Stimulation , Visual Cortex , Male , Female , Mice , Animals , Thalamus/physiology , Neuronal Plasticity/physiology , Visual Cortex/physiology , Synapses
3.
J Neurosci ; 42(42): 7921-7930, 2022 10 19.
Article in English | MEDLINE | ID: mdl-36261269

ABSTRACT

Sensory loss leads to widespread cross-modal plasticity across brain areas to allow the remaining senses to guide behavior. While multimodal sensory interactions are often attributed to higher-order sensory areas, cross-modal plasticity has been observed at the level of synaptic changes even across primary sensory cortices. In particular, vision loss leads to widespread circuit adaptation in the primary auditory cortex (A1) even in adults. Here we report using mice of both sexes in which cross-modal plasticity occurs even earlier in the sensory-processing pathway at the level of the thalamus in a modality-selective manner. A week of visual deprivation reduced inhibitory synaptic transmission from the thalamic reticular nucleus (TRN) to the primary auditory thalamus (MGBv) without changes to the primary visual thalamus (dLGN). The plasticity of TRN inhibition to MGBv was observed as a reduction in postsynaptic gain and short-term depression. There was no observable plasticity of the cortical feedback excitatory synaptic transmission from the primary visual cortex to dLGN or TRN and A1 to MGBv, which suggests that the visual deprivation-induced plasticity occurs predominantly at the level of thalamic inhibition. We provide evidence that visual deprivation-induced change in the short-term depression of TRN inhibition to MGBv involves endocannabinoid CB1 receptors. TRN inhibition is considered critical for sensory gating, selective attention, and multimodal performances; hence, its plasticity has implications for sensory processing. Our results suggest that selective disinhibition and altered short-term dynamics of TRN inhibition in the spared thalamic nucleus support cross-modal plasticity in the adult brain.SIGNIFICANCE STATEMENT Losing vision triggers adaptation of the brain to enhance the processing of the remaining senses, which can be observed as better auditory performance in blind subjects. We previously found that depriving vision of adult rodents produces widespread circuit reorganization in the primary auditory cortex and enhances auditory processing at a neural level. Here we report that visual deprivation-induced plasticity in adults occurs much earlier in the auditory pathway, at the level of thalamic inhibition. Sensory processing is largely gated at the level of the thalamus via strong cortical feedback inhibition mediated through the thalamic reticular nucleus (TRN). We found that TRN inhibition of the auditory thalamus is selectively reduced by visual deprivation, thus playing a role in adult cross-modal plasticity.


Subject(s)
Endocannabinoids , Thalamic Nuclei , Male , Female , Mice , Animals , Thalamic Nuclei/physiology , Thalamus/physiology , Auditory Pathways/physiology , Synaptic Transmission/physiology
4.
J Neurosci ; 39(20): 3897-3905, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30867257

ABSTRACT

It is well established across many species that neurons in the primary visual cortex (V1) display preference for visual input from one eye or the other, which is termed ocular dominance (OD). In rodents, V1 neurons exhibit a strong bias toward the contralateral eye. Molecular mechanisms of how OD is established and later maintained by plastic changes are largely unknown. Here we report a novel role of an activity-dependent immediate early gene Homer1a (H1a) in these processes. Using both sexes of H1a knock-out (KO) mice, we found that there is basal reduction in the OD index of V1 neurons measured using intrinsic signal imaging. This was because of a reduction in the strength of inputs from the contralateral eye, which is normally dominant in mice. The abnormal basal OD index was not dependent on visual experience and is driven by postnatal expression of H1a. Despite this, H1a KOs still exhibited normal shifts in OD index following a short-term (2-3 d) monocular deprivation (MD) of the contralateral eye with lid suture. However, unlike wild-type counterparts, H1a KOs continued to shift OD index with a longer duration (5-6 d) of MD. The same phenotype was recapitulated in a mouse model that has reduced Homer1 binding to metabotropic glutamate receptor 5 (mGluR5). Our results suggest a novel role of H1a and its interaction with mGluR5 in strengthening contralateral eye inputs during postnatal development to establish normal contralateral bias in mouse V1 without much impact on OD shift with brief MD.SIGNIFICANCE STATEMENT Visual cortical neurons display varying degree of responsiveness to visual stimuli through each eye, which determines their ocular dominance (OD). Molecular mechanisms responsible for establishing normal OD are largely unknown. Development of OD has been shown to be largely independent of visual experience, but guided by molecular cues and spontaneous activity. We found that activity-dependent immediate early gene H1a is critical for establishing normal OD in V1 of mice, which show contralateral eye dominance. Despite the weaker contralateral bias, H1aKOs undergo largely normal OD plasticity. The basic phenotype of H1aKO was recapitulated by mGluR5 mutation that severely reduces H1a interaction. Our results suggest a novel role of mGluR5-H1a interaction in strengthening contralateral eye inputs to V1 during postnatal development.


Subject(s)
Dominance, Ocular/physiology , Homer Scaffolding Proteins/physiology , Neurons/physiology , Visual Cortex/physiology , Animals , Female , Homer Scaffolding Proteins/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Photic Stimulation , Receptor, Metabotropic Glutamate 5/physiology
5.
J Neurosci ; 39(39): 7664-7673, 2019 09 25.
Article in English | MEDLINE | ID: mdl-31413075

ABSTRACT

Homeostatic regulation of synaptic strength allows for maintenance of neural activity within a dynamic range for proper circuit function. There are largely two distinct modes of synaptic plasticity that allow for homeostatic adaptation of cortical circuits: synaptic scaling and sliding threshold (BCM theory). Previous findings suggest that the induction of synaptic scaling is not prevented by blocking NMDARs, whereas the sliding threshold model posits that the synaptic modification threshold of LTP and LTD readjusts with activity and thus the outcome of synaptic plasticity is NMDAR dependent. Although synaptic scaling and sliding threshold have been considered two distinct mechanisms, there are indications from recent studies that these two modes of homeostatic plasticity may interact or that they may operate under two distinct activity regimes. Here, we report using both sexes of mouse that acute genetic knock-out of the obligatory subunit of NMDAR or acute pharmacological block of NMDAR prevents experience-dependent homeostatic regulation of AMPAR-mediated miniature EPSCs in layer 2/3 of visual cortex. This was not due to gross changes in postsynaptic neuronal activity with inhibiting NMDAR function as determine by c-Fos expression and two-photon Ca2+ imaging in awake mice. Our results suggest that experience-dependent homeostatic regulation of intact cortical circuits is mediated by NMDAR-dependent plasticity mechanisms, which supports a sliding threshold model of homeostatic adaptation.SIGNIFICANCE STATEMENT Prolonged changes in sensory experience lead to homeostatic adaptation of excitatory synaptic strength in sensory cortices. Both sliding threshold and synaptic scaling models can account for the observed homeostatic synaptic plasticity. Here we report that visual experience-dependent homeostatic plasticity of excitatory synapses observed in superficial layers of visual cortex is dependent on NMDAR function. In particular, both strengthening of synapses induced by visual deprivation and the subsequent weakening by reinstatement of visual experience were prevented in the absence of functional NMDARs. Our results suggest that sensory experience-dependent homeostatic adaptation depends on NMDARs, which supports the sliding threshold model of plasticity and input-specific homeostatic control observed in vivo.


Subject(s)
Homeostasis/physiology , Neuronal Plasticity/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Visual Cortex/physiology , Animals , Excitatory Postsynaptic Potentials/physiology , Female , Male , Mice , Neurons/physiology
6.
Cereb Cortex ; 29(12): 4919-4931, 2019 12 17.
Article in English | MEDLINE | ID: mdl-30843029

ABSTRACT

Neuropsychiatric disorders share susceptibility genes, suggesting a common origin. One such gene is CNTNAP2 encoding contactin-associated protein 2 (CASPR2), which harbours mutations associated to autism, schizophrenia, and intellectual disability. Antibodies targeting CASPR2 have also been recently described in patients with several neurological disorders, such as neuromyotonia, Morvan's syndrome, and limbic encephalitis. Despite the clear implication of CNTNAP2 and CASPR2 in neuropsychiatric disorders, the pathogenic mechanisms associated with alterations in CASPR2 function are unknown. Here, we show that Caspr2 is expressed in excitatory synapses in the cortex, and that silencing its expression in vitro or in vivo decreases the synaptic expression of α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptors and the amplitude of AMPA receptor-mediated currents. Furthermore, Caspr2 loss of function blocks synaptic scaling in vitro and experience-dependent homoeostatic synaptic plasticity in the visual cortex. Patient CASPR2 antibodies decrease the dendritic levels of Caspr2 and synaptic AMPA receptor trafficking, and perturb excitatory transmission in the visual cortex. These results suggest that mutations in CNTNAP2 may contribute to alterations in AMPA receptor function and homoeostatic plasticity, and indicate that antibodies from anti-CASPR2 encephalitis patients affect cortical excitatory transmission.


Subject(s)
Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/physiology , Receptors, AMPA/metabolism , Synaptic Transmission/physiology , Aged , Animals , Autistic Disorder/genetics , Autoantibodies/immunology , Autoantigens/immunology , Encephalitis/immunology , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Rats , Rats, Wistar , Visual Cortex/metabolism
7.
Nature ; 491(7425): 594-8, 2012 Nov 22.
Article in English | MEDLINE | ID: mdl-23151476

ABSTRACT

The daily solar cycle allows organisms to synchronize their circadian rhythms and sleep-wake cycles to the correct temporal niche. Changes in day-length, shift-work, and transmeridian travel lead to mood alterations and cognitive function deficits. Sleep deprivation and circadian disruption underlie mood and cognitive disorders associated with irregular light schedules. Whether irregular light schedules directly affect mood and cognitive functions in the context of normal sleep and circadian rhythms remains unclear. Here we show, using an aberrant light cycle that neither changes the amount and architecture of sleep nor causes changes in the circadian timing system, that light directly regulates mood-related behaviours and cognitive functions in mice. Animals exposed to the aberrant light cycle maintain daily corticosterone rhythms, but the overall levels of corticosterone are increased. Despite normal circadian and sleep structures, these animals show increased depression-like behaviours and impaired hippocampal long-term potentiation and learning. Administration of the antidepressant drugs fluoxetine or desipramine restores learning in mice exposed to the aberrant light cycle, suggesting that the mood deficit precedes the learning impairments. To determine the retinal circuits underlying this impairment of mood and learning, we examined the behavioural consequences of this light cycle in animals that lack intrinsically photosensitive retinal ganglion cells. In these animals, the aberrant light cycle does not impair mood and learning, despite the presence of the conventional retinal ganglion cells and the ability of these animals to detect light for image formation. These findings demonstrate the ability of light to influence cognitive and mood functions directly through intrinsically photosensitive retinal ganglion cells.


Subject(s)
Affect/radiation effects , Learning/radiation effects , Light , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/radiation effects , Rod Opsins , Affect/drug effects , Affect/physiology , Animals , Antidepressive Agents/pharmacology , Body Temperature Regulation/physiology , Body Temperature Regulation/radiation effects , Circadian Rhythm/physiology , Cognition/drug effects , Cognition/physiology , Cognition/radiation effects , Corticosterone/metabolism , Depression/etiology , Depression/physiopathology , Desipramine/pharmacology , Fluoxetine/pharmacology , Learning/drug effects , Learning/physiology , Long-Term Potentiation/drug effects , Male , Memory/physiology , Memory/radiation effects , Mice , Photoperiod , Retinal Ganglion Cells/drug effects , Rod Opsins/analysis , Sleep/physiology , Wakefulness/physiology
8.
J Neurosci ; 35(23): 8790-801, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-26063913

ABSTRACT

Loss of a sensory modality leads to widespread changes in synaptic function across sensory cortices, which are thought to be the basis for cross-modal adaptation. Previous studies suggest that experience-dependent cross-modal regulation of the spared sensory cortices may be mediated by changes in cortical circuits. Here, we report that loss of vision, in the form of dark exposure (DE) for 1 week, produces laminar-specific changes in excitatory and inhibitory circuits in the primary auditory cortex (A1) of adult mice to promote feedforward (FF) processing and also strengthens intracortical inputs to primary visual cortex (V1). Specifically, DE potentiated FF excitatory synapses from layer 4 (L4) to L2/3 in A1 and recurrent excitatory inputs in A1-L4 in parallel with a reduction in the strength of lateral intracortical excitatory inputs to A1-L2/3. This suggests a shift in processing in favor of FF information at the expense of intracortical processing. Vision loss also strengthened inhibitory synaptic function in L4 and L2/3 of A1, but via laminar specific mechanisms. In A1-L4, DE specifically potentiated the evoked synaptic transmission from parvalbumin-positive inhibitory interneurons to principal neurons without changes in spontaneous miniature IPSCs (mIPSCs). In contrast, DE specifically increased the frequency of mIPSCs in A1-L2/3. In V1, FF excitatory inputs were unaltered by DE, whereas lateral intracortical connections in L2/3 were strengthened, suggesting a shift toward intracortical processing. Our results suggest that loss of vision produces distinct circuit changes in the spared and deprived sensory cortices to shift between FF and intracortical processing to allow adaptation.


Subject(s)
Auditory Cortex/cytology , Nerve Net/physiology , Neural Pathways/physiology , Sensory Deprivation/physiology , Visual Cortex/cytology , Visual Cortex/physiology , Animals , Auditory Cortex/physiology , Channelrhodopsins , Dose-Response Relationship, Drug , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , Female , In Vitro Techniques , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Lysine/analogs & derivatives , Lysine/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Parvalbumins/genetics , Parvalbumins/metabolism , Photic Stimulation , Synapses/physiology , Synaptic Transmission , Red Fluorescent Protein
9.
J Neurosci ; 35(32): 11346-57, 2015 Aug 12.
Article in English | MEDLINE | ID: mdl-26269641

ABSTRACT

Much of the molecular understanding of synaptic pathology in Alzheimer's disease (AD) comes from studies of various mouse models that express familial AD (FAD)-linked mutations, often in combinations. Most studies compare the absolute magnitudes of long-term potentiation (LTP) and long-term depression (LTD) to assess deficits in bidirectional synaptic plasticity accompanying FAD-linked mutations. However, LTP and LTD are not static, but their induction threshold is adjusted by overall neural activity via metaplasticity. Hence LTP/LTD changes in AD mouse models may reflect defects in metaplasticity processes. To determine this, we examined the LTP/LTD induction threshold in APPswe;PS1ΔE9 transgenic (Tg) mice across two different ages. We found that in young Tg mice (1 month), LTP is enhanced at the expense of LTD, but in adults (6 months), the phenotype is reversed to promote LTD and reduce LTP, compared to age-matched wild-type (WT) littermates. The apparent opposite phenotype across age was due to an initial offset in the induction threshold to favor LTP and the inability to undergo developmental metaplasticity in Tg mice. In WTs, the synaptic modification threshold decreased over development to favor LTP and diminish LTD in adults. However, in Tg mice, the magnitudes of LTP and LTD stayed constant across development. The initial offset in LTP/LTD threshold in young Tg mice did not accompany changes in the LTP/LTD induction mechanisms, but altered AMPA receptor phosphorylation and appearance of Ca(2+)-permeable AMPA receptors. We propose that the main synaptic defect in AD mouse models is due to their inability to undergo developmental metaplasticity. SIGNIFICANCE STATEMENT: This work offers a new insight that metaplasticity defects are central to synaptic dysfunctions seen in AD mouse models. In particular, we demonstrate that the apparent differences in LTP/LTD magnitude seen across ages in AD transgenic mouse models reflect the inability to undergo a normal developmental shift in metaplasticity.


Subject(s)
Alzheimer Disease/physiopathology , Hippocampus/physiopathology , Long-Term Potentiation/physiology , Long-Term Synaptic Depression/physiology , Synapses/physiology , Age Factors , Alzheimer Disease/metabolism , Animals , Calcium/metabolism , Disease Models, Animal , Female , Hippocampus/metabolism , Male , Mice , Phosphorylation , Receptors, AMPA/metabolism
10.
J Neurosci ; 34(22): 7575-9, 2014 May 28.
Article in English | MEDLINE | ID: mdl-24872561

ABSTRACT

In primates, the functional connectivity of adult primary visual cortex is susceptible to be modified by sensory training during perceptual learning. It is widely held that this type of neural plasticity might involve mechanisms like long-term potentiation (LTP) and long-term depression (LTD). NMDAR-dependent forms of LTP and LTD are particularly attractive because in rodents they can be induced in a Hebbian manner by near coincidental presynaptic and postsynaptic firing, in a paradigm termed spike timing-dependent plasticity (STDP). These fundamental properties of LTP and LTD, Hebbian induction and NMDAR dependence, have not been examined in primate cortex. Here we demonstrate these properties in the primary visual cortex of the rhesus macaque (Macaca mulatta), and also show that, like in rodents, STDP is gated by neuromodulators. These findings indicate that the cellular principles governing cortical plasticity are conserved across mammalian species, further validating the use of rodents as a model system.


Subject(s)
Long-Term Potentiation/physiology , Neuronal Plasticity/physiology , Synapses/physiology , Visual Cortex/physiology , Animals , Female , Macaca mulatta , Male , Organ Culture Techniques , Synaptic Transmission/physiology
11.
J Neurosci ; 34(32): 10770-9, 2014 Aug 06.
Article in English | MEDLINE | ID: mdl-25100608

ABSTRACT

The developmental increase in the strength of inhibitory synaptic circuits defines the time window of the critical period for plasticity in sensory cortices. Conceptually, plasticity of inhibitory synapses is an attractive mechanism to allow for homeostatic adaptation to the sensory environment. However, a brief duration of visual deprivation that causes maximal change in excitatory synapses produces minimal change in inhibitory synaptic transmission. Here we examined developmental and experience-dependent changes in inhibition by measuring miniature IPSCs (mIPSCs) in layer 2/3 pyramidal neurons of mouse visual cortex. During development from postnatal day 21 (P21) to P35, GABAA receptor function changed from fewer higher-conductance channels to more numerous lower-conductance channels without altering the average mIPSC amplitude. Although a week of visual deprivation did not alter the average mIPSC amplitude, a subsequent 2 h exposure to light produced a rapid rebound potentiation. This form of plasticity is restricted to a critical period before the developmental change in GABAergic synaptic properties is completed, and hence is absent by P35. Visual experience-dependent rebound potentiation of mIPSCs is accompanied by an increase in the open channel number and requires activity-dependent transcription of brain-derived neurotrophic factor (BDNF). Mice lacking BDNF transcription through promoter IV did not show developmental changes in inhibition and lacked rebound potentiation. Our results suggest that sensory experience may have distinct functional consequences in normal versus deprived sensory cortices, and that experience-dependent BDNF expression controls the plasticity of inhibitory synaptic transmission particularly when recovering vision during the critical period.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Neural Inhibition/physiology , Neurons/physiology , Vision, Ocular/physiology , Visual Cortex/cytology , Age Factors , Animals , Animals, Newborn , Biophysics , Brain-Derived Neurotrophic Factor/genetics , Electric Stimulation , Gene Expression Regulation, Developmental/genetics , Glutamate Decarboxylase/metabolism , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Neural Inhibition/drug effects , Neural Inhibition/genetics , Neurons/drug effects , Patch-Clamp Techniques , Sensory Deprivation/physiology
12.
J Neurosci ; 33(31): 12670-8, 2013 Jul 31.
Article in English | MEDLINE | ID: mdl-23904603

ABSTRACT

The impact of aging on cognitive capabilities varies among individuals ranging from significant impairment to preservation of function on par with younger adults. Research on the neural basis for age-related memory decline has focused primarily on the CA1 region of the hippocampus. However, recent studies in elderly human and rodents indicate that individual differences in cognitive aging are more strongly tied to functional alterations in CA3 circuits. To examine synaptic plasticity in the CA3 region, we used aged rats behaviorally characterized in a hippocampal-dependent task to evaluate the status of long-term potentiation and long-term depression (LTP and LTD) in the associative/commissural pathway (A/C → CA3), which provides the majority of excitatory input to CA3 pyramidal neurons. We found that, unlike in CA1 synapses, in A/C → CA3 LTP is minimally affected by age. However, two forms of LTD, involving NMDA and metabotropic glutamate receptors (mGluR), are both greatly reduced in age-impaired rats. Age-unimpaired rats, in contrast, had intact mGluR LTD. These findings indicate that the integrity of mGluR-LTD at A/C → CA3 inputs may play a crucial role in maintaining the performance of CA3 circuitry in aging.


Subject(s)
Aging/physiology , CA3 Region, Hippocampal/cytology , Long-Term Synaptic Depression/physiology , Synapses/physiology , Age Factors , Animals , Biophysics , Cyclopropanes/pharmacology , Electric Stimulation , Excitatory Amino Acid Agents/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , In Vitro Techniques , Long-Term Potentiation/drug effects , Male , Maze Learning/drug effects , Nerve Net/drug effects , Nerve Net/pathology , Neural Inhibition , Pyramidal Cells , Rats , Rats, Long-Evans , Receptors, Metabotropic Glutamate/metabolism , Swimming
13.
J Neurosci ; 33(22): 9306-18, 2013 May 29.
Article in English | MEDLINE | ID: mdl-23719799

ABSTRACT

The tetra(ethylene glycol) derivative of benzothiazole aniline, BTA-EG4, is a novel amyloid-binding small molecule that can penetrate the blood-brain barrier and protect cells from Aß-induced toxicity. However, the effects of Aß-targeting molecules on other cellular processes, including those that modulate synaptic plasticity, remain unknown. We report here that BTA-EG4 decreases Aß levels, alters cell surface expression of amyloid precursor protein (APP), and improves memory in wild-type mice. Interestingly, the BTA-EG4-mediated behavioral improvement is not correlated with LTP, but with increased spinogenesis. The higher dendritic spine density reflects an increase in the number of functional synapses as determined by increased miniature EPSC (mEPSC) frequency without changes in presynaptic parameters or postsynaptic mEPSC amplitude. Additionally, BTA-EG4 requires APP to regulate dendritic spine density through a Ras signaling-dependent mechanism. Thus, BTA-EG4 may provide broad therapeutic benefits for improving neuronal and cognitive function, and may have implications in neurodegenerative disease therapy.


Subject(s)
Aniline Compounds/pharmacology , Benzothiazoles/pharmacology , Dendritic Spines/drug effects , Ethylene Glycols/pharmacology , Genes, ras/drug effects , Neurogenesis/drug effects , Amyloid beta-Protein Precursor/genetics , Animals , Biotinylation , COS Cells , Cerebrovascular Circulation/drug effects , Chlorocebus aethiops , Cognition Disorders/chemically induced , Cognition Disorders/psychology , Enzyme-Linked Immunosorbent Assay , Excitatory Postsynaptic Potentials/drug effects , Immunohistochemistry , Long-Term Potentiation/physiology , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Receptors, AMPA/drug effects
14.
Proc Natl Acad Sci U S A ; 108(33): 13770-5, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21804029

ABSTRACT

The mammalian brain constantly adapts to new experiences of the environment, and inhibitory circuits play a crucial role in this experience-dependent plasticity. A characteristic feature of inhibitory neurons is the establishment of electrical synapses, but the function of electrical coupling in plasticity is unclear. Here we show that elimination of electrical synapses formed by connexin36 altered inhibitory efficacy and caused frequency facilitation of inhibition consistent with a decreased GABA release in the inhibitory network. The altered inhibitory efficacy was paralleled by a failure of theta-burst long-term potentiation induction and by impaired ocular dominance plasticity in the visual cortex. Together, these data suggest a unique mechanism for regulating plasticity in the visual cortex involving synchronization of inhibitory networks via electrical synapses.


Subject(s)
Connexins/physiology , Electrical Synapses , Neuronal Plasticity/physiology , Synaptic Transmission/physiology , Animals , Inhibitory Postsynaptic Potentials , Long-Term Potentiation , Mice , Theta Rhythm , Visual Cortex , gamma-Aminobutyric Acid , Gap Junction delta-2 Protein
15.
Neural Plast ; 2014: 128631, 2014.
Article in English | MEDLINE | ID: mdl-24963413

ABSTRACT

Alzheimer's disease (AD) is the most common form of age-related dementia, which is thought to result from overproduction and/or reduced clearance of amyloid-beta (Aß) peptides. Studies over the past few decades suggest that Aß is produced in an activity-dependent manner and has physiological relevance to normal brain functions. Similarly, physiological functions for ß- and γ-secretases, the two key enzymes that produce Aß by sequentially processing the amyloid precursor protein (APP), have been discovered over recent years. In particular, activity-dependent production of Aß has been suggested to play a role in homeostatic regulation of excitatory synaptic function. There is accumulating evidence that activity-dependent immediate early gene Arc is an activity "sensor," which acts upstream of Aß production and triggers AMPA receptor endocytosis to homeostatically downregulate the strength of excitatory synaptic transmission. We previously reported that Arc is critical for sensory experience-dependent homeostatic reduction of excitatory synaptic transmission in the superficial layers of visual cortex. Here we demonstrate that mice lacking the major neuronal ß-secretase, BACE1, exhibit a similar phenotype: stronger basal excitatory synaptic transmission and failure to adapt to changes in visual experience. Our results indicate that BACE1 plays an essential role in sensory experience-dependent homeostatic synaptic plasticity in the neocortex.


Subject(s)
Amyloid Precursor Protein Secretases/physiology , Aspartic Acid Endopeptidases/physiology , Neuronal Plasticity/physiology , Visual Cortex/physiology , Amyloid Precursor Protein Secretases/genetics , Animals , Aspartic Acid Endopeptidases/genetics , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/physiology , Data Interpretation, Statistical , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , Homeostasis , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Pyramidal Cells/physiology , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Visual Cortex/chemistry
16.
bioRxiv ; 2024 May 03.
Article in English | MEDLINE | ID: mdl-38746420

ABSTRACT

Plastic changes in the brain are primarily limited to early postnatal periods. Recovery of adult brain plasticity is critical for the effective development of therapies. A brief (1-2 week) duration of visual deprivation (dark exposure, DE) in adult mice can trigger functional plasticity of thalamocortical and intracortical circuits in the primary auditory cortex suggesting improved sound processing. We tested if DE enhances the ability of adult mice to detect sounds. We trained and continuously evaluated the behavioral performance of mice in control and DE conditions using automated home-cage training. Consistent with age-related peripheral hearing loss present in C57BL/6J mice, we observed decreased performance for high-frequency sounds with age, which was reduced by DE. In CBA mice with preserved peripheral hearing, we also found that DE enhanced auditory performance in low and mid frequencies over time compared to the control.

17.
J Neurosci ; 32(25): 8469-74, 2012 Jun 20.
Article in English | MEDLINE | ID: mdl-22723686

ABSTRACT

Loss of a sensory modality elicits both unimodal changes in the deprived cortex and cross-modal alterations in the remaining sensory systems. Unimodal changes are proposed to recruit the deprived cortex for processing the remaining senses, while cross-modal changes are thought to refine processing of spared senses. Hence coordinated unimodal and cross-modal changes are likely beneficial. Despite this expectation, we report in mice that losing behaviorally relevant patterned vision is sufficient to trigger cross-modal synaptic changes in the primary somatosensory cortex barrel fields, but is insufficient to drive unimodal synaptic plasticity in visual cortex (V1), which requires a complete loss of visual activity. In addition, cross-modal changes depend on whisker inputs. Our results demonstrate that unimodal and cross-modal synaptic plasticity occur independently of each other and rely on distinct sensory requirements.


Subject(s)
Homeostasis/physiology , Neuronal Plasticity/physiology , Synapses/physiology , Animals , Blindness/physiopathology , Darkness , Excitatory Postsynaptic Potentials/physiology , Eye Enucleation , Eyelids/physiology , Female , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Receptors, AMPA/physiology , Recruitment, Neurophysiological/physiology , Sensory Deprivation/physiology , Somatosensory Cortex/physiology , Vibrissae/innervation , Vision, Ocular/physiology , Visual Cortex/physiology
18.
J Neurosci ; 32(43): 15027-35, 2012 Oct 24.
Article in English | MEDLINE | ID: mdl-23100424

ABSTRACT

Metaplasticity, the adaptive changes of long-term potentiation (LTP) and long-term depression (LTD) in response to fluctuations in neural activity is well documented in visual cortex, where dark rearing shifts the frequency threshold for the induction of LTP and LTD. Here we studied metaplasticity affecting spike-timing-dependent plasticity, in which the polarity of plasticity is determined not by the stimulation frequency, but by the temporal relationship between near-coincidental presynaptic and postsynaptic firing. We found that in mouse visual cortex the same regime of deprivation that restricts the frequency range for inducing rate-dependent LTD extends the integration window for inducing timing-dependent LTD, enabling LTD induction with random presynaptic and postsynaptic firing. Notably, the underlying mechanism for the changes in both rate-dependent and time-dependent LTD appears to be an increase of NR2b-containing NMDAR at the synapse. Thus, the rules of metaplasticity might manifest in opposite directions, depending on the plasticity-induction paradigms.


Subject(s)
Action Potentials/physiology , Darkness , Neuronal Plasticity/physiology , Visual Cortex/cytology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Action Potentials/drug effects , Analysis of Variance , Animals , Biophysics , Electric Stimulation , Excitatory Amino Acid Antagonists/pharmacology , Female , GABA Antagonists/pharmacology , In Vitro Techniques , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Long-Term Synaptic Depression/drug effects , Long-Term Synaptic Depression/physiology , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Patch-Clamp Techniques , Pyridazines/pharmacology , Time Factors
19.
Semin Cell Dev Biol ; 22(5): 514-20, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21856433

ABSTRACT

Discovery of long-term potentiation (LTP) in the dentate gyrus of the rabbit hippocampus by Bliss and Lømo opened up a whole new field to study activity-dependent long-term synaptic modifications in the brain. Since then hippocampal synapses have been a key model system to study the mechanisms of different forms of synaptic plasticity. At least for the postsynaptic forms of LTP and long-term depression (LTD), regulation of AMPA receptors (AMPARs) has emerged as a key mechanism. While many of the synaptic plasticity mechanisms uncovered in at the hippocampal synapses apply to synapses across diverse brain regions, there are differences in the mechanisms that often reveal the specific functional requirements of the brain area under study. Here we will review AMPAR regulation underlying synaptic plasticity in hippocampus and neocortex. The main focus of this review will be placed on postsynaptic forms of synaptic plasticity that impinge on the regulation of AMPARs using hippocampal CA1 and primary sensory cortices as examples. And through the comparison, we will highlight the key similarities and functional differences between the two synapses.


Subject(s)
CA1 Region, Hippocampal/metabolism , Long-Term Potentiation/physiology , Long-Term Synaptic Depression/physiology , Models, Biological , Neocortex/metabolism , Receptors, AMPA/metabolism , CA1 Region, Hippocampal/physiology , Humans , Neocortex/physiology , Receptors, AMPA/physiology , Synapses/metabolism , Synapses/physiology
20.
Nat Commun ; 14(1): 7165, 2023 11 07.
Article in English | MEDLINE | ID: mdl-37935668

ABSTRACT

Somatostatin-positive (SOM) interneurons are integral for shaping cortical processing and their dynamic recruitment is likely necessary for adaptation to sensory experience and contextual information. We found that excitatory synapses on SOMs in layer 2/3 (L2/3) of primary visual cortex (V1) of mice can be categorized into fast (F)- and slow (S)-Types based on the kinetics of the AMPA receptor-mediated current. Each SOM contains both types of synapses in varying proportions. The majority of local pyramidal neurons (PCs) make unitary connections with SOMs using both types, followed by those utilizing only S-Type, and a minority with only F-Type. Sensory experience differentially regulates synapses on SOMs, such that local F-Type synapses change with visual deprivation and S-Type synapses undergo plasticity with crossmodal auditory deprivation. Our results demonstrate that the two types of excitatory synapses add richness to the SOM circuit recruitment and undergo selective plasticity enabling dynamic adaptation of the adult V1.


Subject(s)
Synapses , Visual Cortex , Mice , Animals , Synapses/metabolism , Interneurons/metabolism , Pyramidal Cells/metabolism , Somatostatin/metabolism , Visual Cortex/physiology
SELECTION OF CITATIONS
SEARCH DETAIL