Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Neuroinflammation ; 20(1): 269, 2023 Nov 17.
Article in English | MEDLINE | ID: mdl-37978387

ABSTRACT

Alzheimer's disease (AD) pathology and amyloid-beta (Aß) plaque deposition progress slowly in the cerebellum compared to other brain regions, while the entorhinal cortex (EC) is one of the most vulnerable regions. Using a knock-in AD mouse model (App KI), we show that within the cerebellum, the deep cerebellar nuclei (DCN) has particularly low accumulation of Aß plaques. To identify factors that might underlie differences in the progression of AD-associated neuropathology across regions, we profiled gene expression in single nuclei (snRNAseq) across all cell types in the DCN and EC of wild-type (WT) and App KI male mice at age 7 months. We found differences in expression of genes associated with inflammatory activation, PI3K-AKT signalling, and neuron support functions between both regions and genotypes. In WT mice, the expression of interferon-response genes in microglia is higher in the DCN than the EC and this enrichment is confirmed by RNA in situ hybridisation, and measurement of inflammatory cytokines by protein array. Our analyses also revealed that multiple glial populations are responsible for establishing this cytokine-enriched niche. Furthermore, homogenates derived from the DCN induced inflammatory gene expression in BV2 microglia. We also assessed the relationship between the DCN microenvironment and Aß pathology by depleting microglia using a CSF1R inhibitor PLX5622 and saw that, surprisingly, the expression of a subset of inflammatory cytokines was increased while plaque abundance in the DCN was further reduced. Overall, our study revealed the presence of a cytokine-enriched microenvironment unique to the DCN that when modulated, can alter plaque deposition.


Subject(s)
Alzheimer Disease , Cytokines , Mice , Male , Animals , Cytokines/genetics , Cytokines/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Plaque, Amyloid/pathology , Mice, Transgenic , Cerebellar Nuclei/metabolism , Cerebellar Nuclei/pathology , Phosphatidylinositol 3-Kinases/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Microglia/metabolism , Disease Models, Animal
2.
iScience ; 26(9): 107707, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37694138

ABSTRACT

A single episode of pilocarpine-induced status epilepticus can trigger the development of spontaneous recurrent seizures in a rodent model for epilepsy. The initial seizure-induced events in neuronal nuclei that lead to long-term changes in gene expression and cellular responses likely contribute toward epileptogenesis. Using a transgenic mouse model to specifically isolate excitatory neuronal nuclei, we profiled the seizure-induced nuclear proteome via tandem mass tag mass spectrometry and observed robust enrichment of nuclear proteins associated with the SUMOylation pathway. In parallel with nuclear proteome, we characterized nuclear gene expression by RNA sequencing which provided insights into seizure-driven transcriptional regulation and dynamics. Strikingly, we saw widespread downregulation of zinc-finger transcription factors, specifically proteins that harbor Krüppel-associated box (KRAB) domains. Our results provide a detailed snapshot of nuclear events induced by seizure activity and demonstrate a robust method for cell-type-specific nuclear profiling that can be applied to other cell types and models.

3.
Cereb Cortex ; 33(3): 676-690, 2023 01 05.
Article in English | MEDLINE | ID: mdl-35253866

ABSTRACT

The amygdala is known to modulate hippocampal synaptic plasticity. One role could be an immediate effect of basolateral amygdala (BLA) in priming synaptic plasticity in the hippocampus. Another role could be through associative synaptic co-operation and competition that triggers events involved in the maintenance of synaptic potentiation. We present evidence that the timing and activity level of BLA stimulation are important factors for the induction and maintenance of long-term potentiation (LTP) in ventral hippocampal area CA1. A 100 Hz BLA co-stimulation facilitated the induction of LTP, whereas 200 Hz co-stimulation attenuated induction. A 100 Hz BLA co-stimulation also caused enhanced persistence, sufficient to prevent synaptic competition. This maintenance effect is likely through translational mechanisms, as mRNA expression of primary response genes was unaffected, whereas protein level of plasticity-related products was increased. Further understanding of the neural mechanisms of amygdala modulation on hippocampus could provide insights into the mechanisms of emotional disorders.


Subject(s)
Basolateral Nuclear Complex , Neuronal Plasticity , Neuronal Plasticity/physiology , Hippocampus/physiology , Long-Term Potentiation/physiology , Amygdala/physiology , Electric Stimulation
7.
Nature ; 600(7888): 269-273, 2021 12.
Article in English | MEDLINE | ID: mdl-34789878

ABSTRACT

The brain is the seat of body weight homeostasis. However, our inability to control the increasing prevalence of obesity highlights a need to look beyond canonical feeding pathways to broaden our understanding of body weight control1-3. Here we used a reverse-translational approach to identify and anatomically, molecularly and functionally characterize a neural ensemble that promotes satiation. Unbiased, task-based functional magnetic resonance imaging revealed marked differences in cerebellar responses to food in people with a genetic disorder characterized by insatiable appetite. Transcriptomic analyses in mice revealed molecularly and topographically -distinct neurons in the anterior deep cerebellar nuclei (aDCN) that are activated by feeding or nutrient infusion in the gut. Selective activation of aDCN neurons substantially decreased food intake by reducing meal size without compensatory changes to metabolic rate. We found that aDCN activity terminates food intake by increasing striatal dopamine levels and attenuating the phasic dopamine response to subsequent food consumption. Our study defines a conserved satiation centre that may represent a novel therapeutic target for the management of excessive eating, and underscores the utility of a 'bedside-to-bench' approach for the identification of neural circuits that influence behaviour.


Subject(s)
Body Weight Maintenance/genetics , Body Weight Maintenance/physiology , Cerebellum/physiology , Food , Protein Biosynthesis , Reverse Genetics , Satiety Response/physiology , Adult , Animals , Appetite Regulation/genetics , Appetite Regulation/physiology , Cerebellar Nuclei/cytology , Cerebellar Nuclei/physiology , Cerebellum/cytology , Cues , Dopamine/metabolism , Eating/genetics , Eating/physiology , Feeding Behavior/physiology , Female , Homeostasis , Humans , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Neostriatum/metabolism , Neurons/physiology , Obesity/genetics , Philosophy , Young Adult
8.
Aging Cell ; 20(12): e13502, 2021 12.
Article in English | MEDLINE | ID: mdl-34796608

ABSTRACT

Clinical studies have shown that female brains are more predisposed to neurodegenerative diseases such as Alzheimer's disease (AD), but the cellular and molecular mechanisms behind this disparity remain unknown. In several mouse models of AD, synaptic plasticity dysfunction is an early event and appears before significant accumulation of amyloid plaques and neuronal degeneration. However, it is unclear whether sexual dimorphism at the synaptic level contributes to the higher risk and prevalence of AD in females. Our studies on APP/PS1 (APPSwe/PS1dE9) mouse model show that AD impacts hippocampal long-term plasticity in a sex-specific manner. Long-term potentiation (LTP) induced by strong tetanic stimulation (STET), theta burst stimulation (TBS) and population spike timing-dependent plasticity (pSTDP) show a faster decay in AD females compared with age-matched AD males. In addition, behavioural tagging (BT), a model of associative memory, is specifically impaired in AD females with a faster decay in memory compared with males. Together with the plasticity and behavioural data, we also observed an upregulation of neuroinflammatory markers, along with downregulation of transcripts that regulate cellular processes associated with synaptic plasticity and memory in females. Immunohistochemistry of AD brains confirms that female APP/PS1 mice carry a higher amyloid plaque burden and have enhanced microglial activation compared with male APP/PS1 mice. Their presence in the diseased mice also suggests a link between the impairment of LTP and the upregulation of the inflammatory response. Overall, our data show that synaptic plasticity and associative memory impairments are more prominent in females and this might account for the faster progression of AD in females.


Subject(s)
Alzheimer Disease/physiopathology , Memory Disorders/physiopathology , Neuronal Plasticity/immunology , Animals , Disease Models, Animal , Female , Mice , Sex Factors
9.
Neuromolecular Med ; 22(1): 1-24, 2020 03.
Article in English | MEDLINE | ID: mdl-31346933

ABSTRACT

Regulated intramembrane proteolysis (RIP) occurs in a cell when transmembrane proteins are cleaved by intramembrane proteases such as secretases to generate soluble protein fragments in the extracellular environment and the cytosol. In the cytosol, these soluble intracellular domains (ICDs) have local functions near the site of cleavage or in many cases, translocate to the nucleus to modulate gene expression. While the mechanism of RIP is relatively well studied, the fate and function of ICDs for most substrate proteins remain poorly characterized. In neurons, RIP occurs in various subcellular compartments including at the synapse. In this review, we summarize current research on RIP in neurons, focusing specifically on synaptic proteins where the presence and function of the ICDs have been reported. We also briefly discuss activity-driven processing of RIP substrates at the synapse and the cellular machinery that support long-distance transport of ICDs from the synapse to the nucleus. Finally, we describe future challenges in this field of research in the context of understanding the contribution of ICDs in neuronal function.


Subject(s)
Cell Membrane/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Protein Domains/physiology , Protein Transport/physiology , Active Transport, Cell Nucleus/physiology , Amyloid Precursor Protein Secretases/metabolism , Cytosol/metabolism , Humans , Nerve Tissue Proteins/chemistry , Neurons/ultrastructure , Peptide Hydrolases/metabolism , Protein Processing, Post-Translational , Proteolysis , Solubility , Subcellular Fractions/metabolism , Synapses/metabolism
10.
Neurobiol Learn Mem ; 138: 78-84, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27473670

ABSTRACT

The unique polarity of neurons requires that synaptic inputs are relayed to the nucleus to trigger changes in gene expression. This long distance signaling process is crucial for the function and survival of neuronal circuits. To that end, neurons have developed multiple modes of signal transmission from the synapse to the nucleus. In this review, we summarize the latest research on activity-dependent movement and nuclear import of postsynaptic proteins that modulate neuronal plasticity. We also focus on the mechanism of active transport as well as the role of importins in mediating nuclear import of the postsynaptic proteins. Finally, we briefly discuss the role of synapse to nucleus signaling in the context of transcription-dependent plasticity and conclude by describing future challenges in this field of research.


Subject(s)
Cell Nucleus/metabolism , Neuronal Plasticity/physiology , Signal Transduction/physiology , Synapses/metabolism , Animals , Humans
11.
Front Mol Neurosci ; 8: 48, 2015.
Article in English | MEDLINE | ID: mdl-26388727

ABSTRACT

Previous studies have revealed a critical role for CREB-regulated transcriptional coactivator (CRTC1) in regulating neuronal gene expression during learning and memory. CRTC1 localizes to synapses but undergoes activity-dependent nuclear translocation to regulate the transcription of CREB target genes. Here we investigate the long-distance retrograde transport of CRTC1 in hippocampal neurons. We show that local elevations in calcium, triggered by activation of glutamate receptors and L-type voltage-gated calcium channels, initiate active, dynein-mediated retrograde transport of CRTC1 along microtubules. We identify a nuclear localization signal within CRTC1, and characterize three conserved serine residues whose dephosphorylation is required for nuclear import. Domain analysis reveals that the amino-terminal third of CRTC1 contains all of the signals required for regulated nucleocytoplasmic trafficking. We fuse this region to Dendra2 to generate a reporter construct and perform live-cell imaging coupled with local uncaging of glutamate and photoconversion to characterize the dynamics of stimulus-induced retrograde transport and nuclear accumulation.

12.
J Virol ; 87(17): 9431-40, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23804637

ABSTRACT

Alphaherpesviruses, including pseudorabies virus (PRV), spread directionally within the nervous systems of their mammalian hosts. Three viral membrane proteins are required for efficient anterograde-directed spread of infection in neurons, including Us9 and a heterodimer composed of the glycoproteins gE and gI. We previously demonstrated that the kinesin-3 motor KIF1A mediates anterograde-directed transport of viral particles in axons of cultured peripheral nervous system (PNS) neurons. The PRV Us9 protein copurifies with KIF1A, recruiting the motor to transport vesicles, but at least one unidentified additional viral protein is necessary for this interaction. Here we show that gE/gI are required for efficient anterograde transport of viral particles in axons by mediating the interaction between Us9 and KIF1A. In the absence of gE/gI, viral particles containing green fluorescent protein (GFP)-tagged Us9 are assembled in the cell body but are not sorted efficiently into axons. Importantly, we found that gE/gI are necessary for efficient copurification of KIF1A with Us9, especially at early times after infection. We also constructed a PRV recombinant that expresses a functional gE-GFP fusion protein and used affinity purification coupled with mass spectrometry to identify gE-interacting proteins. Several viral and host proteins were found to associate with gE-GFP. Importantly, both gI and Us9, but not KIF1A, copurified with gE-GFP. We propose that gE/gI are required for efficient KIF1A-mediated anterograde transport of viral particles because they indirectly facilitate or stabilize the interaction between Us9 and KIF1A.


Subject(s)
Alphaherpesvirinae/physiology , Herpesvirus 1, Suid/physiology , Kinesins/physiology , Lipoproteins/physiology , Neurons/physiology , Neurons/virology , Phosphoproteins/physiology , Viral Envelope Proteins/physiology , Viral Proteins/physiology , Alphaherpesvirinae/genetics , Alphaherpesvirinae/pathogenicity , Animals , Axonal Transport/physiology , Cell Line , Cells, Cultured , Herpesvirus 1, Suid/genetics , Herpesvirus 1, Suid/pathogenicity , Host-Pathogen Interactions , Intracellular Signaling Peptides and Proteins , Lipoproteins/genetics , PC12 Cells , Phosphoproteins/genetics , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Swine , Viral Envelope Proteins/genetics , Viral Proteins/genetics , Virion/physiology
13.
Cell ; 150(1): 207-21, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22770221

ABSTRACT

Long-lasting changes in synaptic efficacy, such as those underlying long-term memory, require transcription. Activity-dependent transport of synaptically localized transcriptional regulators provides a direct means of coupling synaptic stimulation with changes in transcription. The CREB-regulated transcriptional coactivator (CRTC1), which is required for long-term hippocampal plasticity, binds CREB to potently promote transcription. We show that CRTC1 localizes to synapses in silenced hippocampal neurons but translocates to the nucleus in response to localized synaptic stimulation. Regulated nuclear translocation occurs only in excitatory neurons and requires calcium influx and calcineurin activation. CRTC1 is controlled in a dual fashion with activity regulating CRTC1 nuclear translocation and cAMP modulating its persistence in the nucleus. Neuronal activity triggers a complex change in CRTC1 phosphorylation, suggesting that CRTC1 may link specific types of stimuli to specific changes in gene expression. Together, our results indicate that synapse-to-nuclear transport of CRTC1 dynamically informs the nucleus about synaptic activity.


Subject(s)
Cell Nucleus/metabolism , Hippocampus/cytology , Neurons/metabolism , Synapses/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , Hippocampus/metabolism , Mice , Protein Transport , Rats
14.
Curr Opin Neurobiol ; 21(2): 345-52, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21349698

ABSTRACT

Signals generated in distal subcellular compartments of neurons must often travel long distances to the nucleus to trigger changes in gene expression. This retrograde signaling is critical to the development, function, and survival of neural circuits, and neurons have evolved multiple mechanisms to transmit signals over long distances. In this review, we briefly summarize the range of mechanisms whereby distally generated signals are transported to neuronal nuclei. We then focus on the transport of soluble signals from the synapse to the nucleus during neuronal plasticity.


Subject(s)
Cell Nucleus/metabolism , Neuronal Plasticity/physiology , Neurons/metabolism , Signal Transduction/physiology , Synapses/metabolism , Animals , Humans
15.
Mol Cell Neurosci ; 43(1): 108-16, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19837165

ABSTRACT

The Ras effector Rin1 is induced concomitant with synaptogenesis in forebrain neurons, where it inhibits fear conditioning and amygdala LTP. In epithelial cells, lower levels of Rin1 orchestrate receptor endocytosis. A 945 bp Rin1 promoter fragment was active in hippocampal neurons and directed accurate tissue-specific and temporal expression in transgenic mice. Regulated expression in neurons and epithelial cells was mediated in part by Snail transcriptional repressors: mutation of a conserved Snail site increased expression and endogenous Snai1 was detected at the Rin1 promoter. We also describe an element closely related to, but distinct from, the consensus site for REST, a master repressor of neuronal genes. Conversion to a consensus REST sequence reduced expression in both cell types. These results provide insight into regulated expression of a neuronal Ras effector, define a promoter useful in telencephalic neuron studies, and describe a novel REST site variant directing expression to mature neurons.


Subject(s)
Gene Expression Regulation , Neurons/physiology , Prosencephalon/cytology , rab GTP-Binding Proteins/metabolism , Animals , Cells, Cultured , Genes, Reporter , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Transgenic , Neurons/cytology , Promoter Regions, Genetic , Prosencephalon/metabolism , Signal Transduction/physiology , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , rab GTP-Binding Proteins/genetics
16.
J Neurosci ; 29(50): 15613-20, 2009 Dec 16.
Article in English | MEDLINE | ID: mdl-20016075

ABSTRACT

Synaptic plasticity, the capacity of neurons to change the strength of their connections with experience, provides a mechanism for learning and memory in the brain. Long-term plasticity requires new transcription, indicating that synaptically generated signals must be transported to the nucleus. Previous studies have described a role for importin nuclear transport adaptors in mediating the retrograde transport of signals from synapse to nucleus during plasticity. Here, we investigated the possibility that stimulus-induced translocation of importins from synapse to nucleus involves activity-dependent anchoring of importins at the synapse. We show that importin alpha binds to a nuclear localization signal (NLS) present in the cytoplasmic tail of NR1-1a. This interaction is disrupted by activation of NMDA receptors in cultured neurons and by stimuli that trigger late-phase, but not early-phase, long-term potentiation of CA3-CA1 synapses in acute hippocampal slices. In vitro PKC phosphorylation of GST-NR1-1a abolishes its ability to bind importin alpha in brain lysates, and the interaction of importin alpha and NR1 in neurons is modulated by PKC activity. Together, our results indicate that importin alpha is tethered at the postsynaptic density by binding to the NLS present in NR1-1a. This interaction is activity dependent, with importin alpha being released following NMDA receptor activation and phosphorylation rendering it available to bind soluble cargoes and transport them to the nucleus during transcription-dependent forms of neuronal plasticity.


Subject(s)
Cytoplasm/metabolism , Protein Subunits/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , alpha Karyopherins/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Cells, Cultured , Cytoplasm/genetics , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Protein Binding/physiology , Protein Subunits/genetics , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics , Synapses/genetics , alpha Karyopherins/genetics
17.
Curr Protoc Cell Biol ; Chapter 26: Unit 26.4, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19499506

ABSTRACT

Compartmented neuronal cultures allow experimenters to establish separate fluid environments for neuronal axons and the soma from which they emanate. Physical isolation of cell bodies and axons is achieved by culturing neurons in tri-chambered Teflon rings. Dissociated ganglia are plated in one end compartment of the trichamber, and axonal growth is guided underneath watertight silicone grease barriers into a separate compartment. Since the axons and cell bodies are located in different compartments, they can be infected and assayed separately. We describe the assembly and use of compartmented neuronal cultures for in vitro study of directional infection of neurons by alpha herpesviruses. Selective application of viral inoculum to only one compartment ensures that the remainder of the neuron is not contaminated by input inoculum. This allows for quantification of viral spread, and unambiguous interpretation of immunofluorescence and electron microscopy images.


Subject(s)
Cell Culture Techniques/methods , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Herpesviridae/physiology , Neurons/pathology , Neurons/virology , Animals , Embryo, Mammalian , Female , Fluorescent Antibody Technique , Ganglia, Sympathetic , Guided Tissue Regeneration , Herpesviridae/pathogenicity , Herpesviridae Infections/physiopathology , Mice , Microscopy, Electron , Neurons/metabolism , Pregnancy , Rats , Virulence
18.
Proc Natl Acad Sci U S A ; 105(44): 17175-80, 2008 Nov 04.
Article in English | MEDLINE | ID: mdl-18957537

ABSTRACT

Signals received at distal synapses of neurons must be conveyed to the nucleus to initiate the changes in transcription that underlie long-lasting synaptic plasticity. The presence of importin nuclear transporters and of select transcription factors at synapses raises the possibility that importins directly transport transcription factors from synapse to nucleus to modulate gene expression. Here, we show that cyclic AMP response element binding protein 2 (CREB2)/activating transcription factor 4 (ATF4), a transcriptional repressor that modulates long-term synaptic plasticity and memory, localizes to distal dendrites of rodent hippocampal neurons and neurites of Aplysia sensory neurons (SNs) and binds to specific importin alpha isoforms. Binding of CREB2 to importin alpha is required for its transport from distal dendrites to the soma and for its translocation into the nucleus. CREB2 accumulates in the nucleus during long-term depression (LTD) but not long-term potentiation of rodent hippocampal synapses, and during LTD but not long-term facilitation (LTF) of Aplysia sensory-motor synapses. Time-lapse microscopy of CREB2 tagged with a photoconvertible fluorescent protein further reveals retrograde transport of CREB2 from distal neurites to the nucleus of Aplysia SN during phenylalanine-methionine-arginine-phenylalanine-amide (FMRFamide)-induced LTD. Together, our findings indicate that CREB2 is a novel cargo of importin alpha that translocates from distal synaptic sites to the nucleus after stimuli that induce LTD of neuronal synapses.


Subject(s)
Cell Nucleus/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Nerve Tissue Proteins/metabolism , Repressor Proteins/metabolism , Sensory Receptor Cells/metabolism , alpha Karyopherins/metabolism , Active Transport, Cell Nucleus , Animals , Cell Culture Techniques , FMRFamide/pharmacology , Fluorescent Antibody Technique , Long-Term Potentiation , Long-Term Synaptic Depression , Presynaptic Terminals , Rats , Transfection
19.
Methods Mol Biol ; 292: 299-316, 2005.
Article in English | MEDLINE | ID: mdl-15507717

ABSTRACT

This chapter discusses the culture of primary sympathetic neurons (superior cervical ganglia) from rat embryos and PC12 cells differentiated into neurons for use in viral infection experiments. Methods are described for the use of a neurotropic herpesvirus, pseudorabies virus (PRV), to analyze the assembly, egress, and transport of viral antigens in neurons.


Subject(s)
Clinical Laboratory Techniques , Herpesvirus 1, Suid/metabolism , Neurons/virology , Pseudorabies/metabolism , Animals , Cell Culture Techniques/methods , Embryo, Mammalian/cytology , Neurons/metabolism , PC12 Cells , Rats , Superior Cervical Ganglion/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...