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.
J Biomed Sci ; 31(1): 69, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992696

ABSTRACT

BACKGROUND: Local translation at synapses is important for rapidly remodeling the synaptic proteome to sustain long-term plasticity and memory. While the regulatory mechanisms underlying memory-associated local translation have been widely elucidated in the postsynaptic/dendritic region, there is no direct evidence for which RNA-binding protein (RBP) in axons controls target-specific mRNA translation to promote long-term potentiation (LTP) and memory. We previously reported that translation controlled by cytoplasmic polyadenylation element binding protein 2 (CPEB2) is important for postsynaptic plasticity and memory. Here, we investigated whether CPEB2 regulates axonal translation to support presynaptic plasticity. METHODS: Behavioral and electrophysiological assessments were conducted in mice with pan neuron/glia- or glutamatergic neuron-specific knockout of CPEB2. Hippocampal Schaffer collateral (SC)-CA1 and temporoammonic (TA)-CA1 pathways were electro-recorded to monitor synaptic transmission and LTP evoked by 4 trains of high-frequency stimulation. RNA immunoprecipitation, coupled with bioinformatics analysis, were used to unveil CPEB2-binding axonal RNA candidates associated with learning, which were further validated by Western blotting and luciferase reporter assays. Adeno-associated viruses expressing Cre recombinase were stereotaxically delivered to the pre- or post-synaptic region of the TA circuit to ablate Cpeb2 for further electrophysiological investigation. Biochemically isolated synaptosomes and axotomized neurons cultured on a microfluidic platform were applied to measure axonal protein synthesis and FM4-64FX-loaded synaptic vesicles. RESULTS: Electrophysiological analysis of hippocampal CA1 neurons detected abnormal excitability and vesicle release probability in CPEB2-depleted SC and TA afferents, so we cross-compared the CPEB2-immunoprecipitated transcriptome with a learning-induced axonal translatome in the adult cortex to identify axonal targets possibly regulated by CPEB2. We validated that Slc17a6, encoding vesicular glutamate transporter 2 (VGLUT2), is translationally upregulated by CPEB2. Conditional knockout of CPEB2 in VGLUT2-expressing glutamatergic neurons impaired consolidation of hippocampus-dependent memory in mice. Presynaptic-specific ablation of Cpeb2 in VGLUT2-dominated TA afferents was sufficient to attenuate protein synthesis-dependent LTP. Moreover, blocking activity-induced axonal Slc17a6 translation by CPEB2 deficiency or cycloheximide diminished the releasable pool of VGLUT2-containing synaptic vesicles. CONCLUSIONS: We identified 272 CPEB2-binding transcripts with altered axonal translation post-learning and established a causal link between CPEB2-driven axonal synthesis of VGLUT2 and presynaptic translation-dependent LTP. These findings extend our understanding of memory-related translational control mechanisms in the presynaptic compartment.


Subject(s)
Neuronal Plasticity , RNA-Binding Proteins , Synaptic Transmission , Vesicular Glutamate Transport Protein 2 , Animals , Mice , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Neuronal Plasticity/physiology , Synaptic Transmission/physiology , Vesicular Glutamate Transport Protein 2/metabolism , Vesicular Glutamate Transport Protein 2/genetics , Mice, Knockout , Axons/metabolism , Axons/physiology , RNA, Messenger/metabolism , RNA, Messenger/genetics , Male , Protein Biosynthesis
2.
Trends Genet ; 38(10): 991-995, 2022 10.
Article in English | MEDLINE | ID: mdl-35637074

ABSTRACT

Polyploidization and polyploidy reversal (depolyploidization) are crucial pathways to conversely alter genomic contents in organisms. Understanding the mechanisms switching between polyploidization and polyploidy reversal should broaden our knowledge of the generation of pathological polyploidy and pave a new path to prevent related diseases.


Subject(s)
Mitosis , Polyploidy , Humans
3.
Neuropsychopharmacology ; 46(9): 1669-1679, 2021 08.
Article in English | MEDLINE | ID: mdl-33941859

ABSTRACT

Susceptibility or resilience to posttraumatic stress disorder (PTSD) depends on one's ability to appropriately adjust synaptic plasticity for coping with the traumatic experience. Activity-regulated mRNA translation synthesizes plasticity-related proteins to support long-term synaptic changes and memory. Hence, cytoplasmic polyadenylation element-binding protein 3-knockout (CPEB3-KO) mice, showing dysregulated translation-associated synaptic rigidity, may be susceptible to PTSD-like behavior. Here, using a context-dependent auditory fear conditioning and extinction paradigm, we found that CPEB3-KO mice exhibited traumatic intensity-dependent PTSD-like fear memory. A genome-wide screen of CPEB3-bound transcripts revealed that Nr3c1, encoding glucocorticoid receptor (GR), was translationally suppressed by CPEB3. Thus, CPEB3-KO neurons with elevated GR expression exhibited increased corticosterone-induced calcium influx and decreased mRNA and protein levels of brain-derived neurotrophic factor (Bdnf). Moreover, the reduced expression of BDNF was associated with increased GR level during fear extinction in CPEB3-KO hippocampi. Intracerebroventricular delivery of BDNF before extinction training mitigated spontaneous fear intrusion in CPEB3-KO mice during extinction recall. Analysis of two GEO datasets revealed decreased transcriptomic expression of CPEB3 but not NR3C1 in peripheral blood mononuclear cells of humans with PTSD. Collectively, this study reveals that CPEB3, as a potential PTSD-risk gene, downregulates Nr3c1 translation to maintain proper GR-BDNF signaling for fear extinction.


Subject(s)
Fear , Stress Disorders, Post-Traumatic , Animals , Extinction, Psychological , Leukocytes, Mononuclear , Mice , Mice, Knockout , Protein Biosynthesis , RNA-Binding Proteins , Receptors, Glucocorticoid/genetics
4.
Front Pediatr ; 8: 612279, 2020.
Article in English | MEDLINE | ID: mdl-33634052

ABSTRACT

Brown adipose tissue (BAT) is a thermogenic organ owing to its unique expression of uncoupling protein 1 (UCP1), which is a proton channel in the inner mitochondrial membrane used to dissipate the proton gradient and uncouple the electron transport chain to generate heat instead of adenosine triphosphate. The discovery of metabolically active BAT in human adults, especially in lean people after cold exposure, has provoked the "thermogenic anti-obesity" idea to battle weight gain. Because BAT can expend energy through UCP1-mediated thermogenesis, the molecular mechanisms regulating UCP1 expression have been extensively investigated at both transcriptional and posttranscriptional levels. Of note, the 3'-untranslated region (3'-UTR) of Ucp1 mRNA is differentially processed between mice and humans that quantitatively affects UCP1 synthesis and thermogenesis. Here, we summarize the regulatory mechanisms underlying UCP1 expression, report the number of poly(A) signals identified or predicted in Ucp1 genes across species, and discuss the potential and caution in targeting UCP1 for enhancing thermogenesis and metabolic fitness.

5.
Curr Opin Neurobiol ; 48: 64-70, 2018 02.
Article in English | MEDLINE | ID: mdl-29125978

ABSTRACT

Activity-regulated protein synthesis, especially in the restricted synaptic domains, is critical to maintaining connections and communication between neurons. Accumulating evidence has linked dysregulated translation to various neurodevelopmental or neurodegenerative diseases. In the past 3 decades, after finding ribosomes and specific mRNAs localized around synapses, a significant amount of work has furthered our understanding of how the genetic sequences in mRNAs and their cognate RNA-binding proteins are coordinated to build up synaptic proteomes. Recent exciting findings of various RNA modifications, specialized ribosomes, and their regulatory roles in translation have led to emerging needs to unravel how synaptic ribosomes interpret these hidden codes to regulate molecular connectomes.


Subject(s)
Neurodevelopmental Disorders , Neurons/physiology , RNA-Binding Proteins/metabolism , Ribosomes/physiology , Transcriptome/physiology , Animals , Humans , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/pathology , RNA-Binding Proteins/genetics
6.
Cell Rep ; 21(7): 1783-1794, 2017 Nov 14.
Article in English | MEDLINE | ID: mdl-29141213

ABSTRACT

Activity-dependent synthesis of plasticity-related proteins is necessary to sustain long-lasting synaptic modifications and consolidate memory. We investigated the role of the translational regulator cytoplasmic polyadenylation element binding protein 2 (CPEB2) in learning and memory because regulated mRNA translation contributes to synaptic plasticity. Forebrain-restricted CPEB2 conditional knockout (cKO) mice exhibited impaired hippocampus-dependent memory in contextual fear conditioning and Morris water maze tests. CPEB2 cKO hippocampi showed impaired long-term potentiation in the Schaffer collateral-CA1 pathway. Reduced surface, but not total, expression of AMPA receptors (AMPARs) in CPEB2 KO neurons led us to identify that CPEB2 enhanced the translation of GRASP1 mRNA to facilitate recycling and maintain the surface level of AMPARs. Ectopic expression of CPEB2 or GRASP1 in CA1 areas of CPEB2 cKO mouse hippocampi rescued long-term potentiation and spatial memory in a water maze test. Thus, CPEB2-regulated GRASP1 mRNA translation is pivotal for AMPAR recycling, long-term plasticity, and memory.


Subject(s)
Carrier Proteins/metabolism , Long-Term Potentiation , Maze Learning , RNA-Binding Proteins/metabolism , Receptors, AMPA/metabolism , Animals , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiology , Carrier Proteins/genetics , Cell Membrane/metabolism , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neurons/physiology , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Synapses/metabolism , Synapses/physiology
7.
J Neurosci ; 33(43): 17008-22, 2013 Oct 23.
Article in English | MEDLINE | ID: mdl-24155305

ABSTRACT

Long-term memory requires activity-dependent synthesis of plasticity-related proteins (PRPs) to strengthen synaptic efficacy and consequently consolidate memory. Cytoplasmic polyadenylation element binding protein (CPEB)3 is a sequence-specific RNA-binding protein that regulates translation of several PRP RNAs in neurons. To understand whether CPEB3 plays a part in learning and memory, we generated CPEB3 knock-out (KO) mice and found that the null mice exhibited enhanced hippocampus-dependent, short-term fear memory in the contextual fear conditioning test and long-term spatial memory in the Morris water maze. The basal synaptic transmission of Schaffer collateral-CA1 neurons was normal but long-term depression evoked by paired-pulse low-frequency stimulation was modestly facilitated in the juvenile KO mice. Molecular and cellular characterizations revealed several molecules in regulating plasticity of glutamatergic synapses are translationally elevated in the CPEB3 KO neurons, including the scaffolding protein PSD95 and the NMDA receptors along with the known CPEB3 target, GluA1. Together, CPEB3 functions as a negative regulator to confine the strength of glutamatergic synapses by downregulating the expression of multiple PRPs and plays a role underlying certain forms of hippocampus-dependent memories.


Subject(s)
Guanylate Kinases/metabolism , Hippocampus/physiology , Membrane Proteins/metabolism , Memory, Short-Term , RNA-Binding Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Conditioning, Classical , Disks Large Homolog 4 Protein , Fear , Guanylate Kinases/genetics , Hippocampus/cytology , Hippocampus/metabolism , Long-Term Synaptic Depression , Maze Learning , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Neurons/physiology , RNA-Binding Proteins/genetics , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Synapses/physiology , Synaptic Transmission
8.
J Neurosci Res ; 91(5): 694-705, 2013 May.
Article in English | MEDLINE | ID: mdl-23404572

ABSTRACT

Peripheral injection with a high dose of valproic acid (VPA), a histone deacetylase (HDAC) inhibitor, into animals with mild or moderate spinal cord injury (SCI) for 1 week can reduce spinal cord tissue loss and promote hindlimb locomotor recovery. A purinergic adenosine triphosphate (ATP) receptor subtype, P2X4 receptor (P2X4 R), has been considered as a potential target to diminish SCI-associated inflammatory responses. In this study, using a minipump-based infusion system, we found that intraspinal infusion with VPA for 3 days into injured spinal cord significantly improved hindlimb locomotion of rats with severe SCI induced by a 10-g NYU impactor dropping from the height of 50 mm onto the spinal T9/10 segment. The neuronal fibers in the injured spinal cord tissues were significantly preserved in VPA-treated rats compared with those observed in vehicle-treated animals. Moreover, the accumulation of microglia/macrophages and astrocytes in the injured spinal cord was attenuated in the animal group receiving VPA infusion. VPA also significantly reduced P2X4 R expression post-SCI. Furthermore, in vitro study indicated that VPA, but not the other HDAC inhibitors, sodium butyrate and trichostatin A (TSA), caused downregulation of P2X4 R in microglia activated with lipopolysaccharide (LPS). Moreover, p38 mitogen-activated protein kinase (MAPK)-triggered signaling was involved in the effect of VPA on the inhibition of P2X4 R gene expression. In addition to the findings from others, our results also provide important evidence to show the inhibitory effect of VPA on P2X4 R expression in activated microglia, which may contribute to reduction of SCI-induced gliosis and subsequently preservation of spinal cord tissues. © 2013 Wiley Periodicals, Inc.


Subject(s)
Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Microglia/drug effects , Receptors, Purinergic P2X4/metabolism , Spinal Cord Injuries/pathology , Valproic Acid/pharmacology , Animals , Catalase/metabolism , Cells, Cultured , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Female , Glial Fibrillary Acidic Protein/metabolism , Hindlimb/physiopathology , Locomotion/drug effects , Macrophages/drug effects , Macrophages/pathology , Nerve Fibers/metabolism , Neurofilament Proteins/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X4/genetics , Spinal Cord Injuries/drug therapy , Superoxide Dismutase/metabolism , Valproic Acid/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...