Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 12274, 2024 05 28.
Article in English | MEDLINE | ID: mdl-38806540

ABSTRACT

Cranial irradiation used to control brain malignancies invariably leads to progressive and debilitating declines in cognition. Clinical efforts implementing hippocampal avoidance and NMDAR antagonism, have sought to minimize dose to radiosensitive neurogenic regions while normalizing excitatory/inhibitory (E/I) tone. Results of these trials have yielded only marginal benefits to cognition, prompting current studies to evaluate the potential of systemic extracellular vesicle (EV) therapy to restore neurocognitive functionality in the irradiated brain. Here we tested the hypothesis that EVs derived from inhibitory but not excitatory neuronal cultures would prove beneficial to cognition and associated pathology. Rats subjected to a clinically relevant, fractionated cranial irradiation paradigm were given multiple injections of either GABAergic- or glutamatergic-derived EV and subjected to behavioral testing. Rats treated with GABAergic but not glutamatergic EVs showed significant improvements on hippocampal- and cortical-dependent behavioral tasks. While each treatment enhanced levels of the neurotrophic factors BDNF and GDNF, only GABAergic EVs preserved granule cell neuron dendritic spine density. Additional studies conducted with GABAergic EVs, confirmed significant benefits on amygdala-dependent behavior and modest changes in synaptic plasticity as measured by long-term potentiation. These data point to a potentially more efficacious approach for resolving radiation-induced neurological deficits, possibly through a mechanism able to restore homeostatic E/I balance.


Subject(s)
Cranial Irradiation , Extracellular Vesicles , GABAergic Neurons , Animals , Extracellular Vesicles/metabolism , Rats , Cranial Irradiation/adverse effects , GABAergic Neurons/metabolism , GABAergic Neurons/radiation effects , Male , Hippocampus/radiation effects , Hippocampus/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Neurons/radiation effects , Neurons/metabolism , Glutamic Acid/metabolism , Neuronal Plasticity/radiation effects , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Behavior, Animal/radiation effects
2.
Nat Commun ; 15(1): 3836, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714691

ABSTRACT

Exercise has beneficial effects on cognition throughout the lifespan. Here, we demonstrate that specific exercise patterns transform insufficient, subthreshold training into long-term memory in mice. Our findings reveal a potential molecular memory window such that subthreshold training within this window enables long-term memory formation. We performed RNA-seq on dorsal hippocampus and identify genes whose expression correlate with conditions in which exercise enables long-term memory formation. Among these genes we found Acvr1c, a member of the TGF ß family. We find that exercise, in any amount, alleviates epigenetic repression at the Acvr1c promoter during consolidation. Additionally, we find that ACVR1C can bidirectionally regulate synaptic plasticity and long-term memory in mice. Furthermore, Acvr1c expression is impaired in the aging human and mouse brain, as well as in the 5xFAD mouse model, and over-expression of Acvr1c enables learning and facilitates plasticity in mice. These data suggest that promoting ACVR1C may protect against cognitive impairment.


Subject(s)
Activin Receptors, Type I , Epigenesis, Genetic , Hippocampus , Memory, Long-Term , Physical Conditioning, Animal , Animals , Female , Humans , Male , Mice , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Aging/genetics , Aging/physiology , Hippocampus/metabolism , Memory, Long-Term/physiology , Mice, Inbred C57BL , Neuronal Plasticity/genetics , Physical Conditioning, Animal/physiology , Promoter Regions, Genetic
3.
Alzheimers Dement ; 20(4): 2922-2942, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38460121

ABSTRACT

INTRODUCTION: The BIN1 coding variant rs138047593 (K358R) is linked to Late-Onset Alzheimer's Disease (LOAD) via targeted exome sequencing. METHODS: To elucidate the functional consequences of this rare coding variant on brain amyloidosis and neuroinflammation, we generated BIN1K358R knock-in mice using CRISPR/Cas9 technology. These mice were subsequently bred with 5xFAD transgenic mice, which serve as a model for Alzheimer's pathology. RESULTS: The presence of the BIN1K358R variant leads to increased cerebral amyloid deposition, with a dampened response of astrocytes and oligodendrocytes, but not microglia, at both the cellular and transcriptional levels. This correlates with decreased neurofilament light chain in both plasma and brain tissue. Synaptic densities are significantly increased in both wild-type and 5xFAD backgrounds homozygous for the BIN1K358R variant. DISCUSSION: The BIN1 K358R variant modulates amyloid pathology in 5xFAD mice, attenuates the astrocytic and oligodendrocytic responses to amyloid plaques, decreases damage markers, and elevates synaptic densities. HIGHLIGHTS: BIN1 rs138047593 (K358R) coding variant is associated with increased risk of LOAD. BIN1 K358R variant increases amyloid plaque load in 12-month-old 5xFAD mice. BIN1 K358R variant dampens astrocytic and oligodendrocytic response to plaques. BIN1 K358R variant decreases neuronal damage in 5xFAD mice. BIN1 K358R upregulates synaptic densities and modulates synaptic transmission.


Subject(s)
Alzheimer Disease , Animals , Mice , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides , Disease Models, Animal , Mice, Transgenic , Neuroglia/pathology , Plaque, Amyloid/pathology , Humans
4.
Cancer Res Commun ; 3(4): 725-737, 2023 04.
Article in English | MEDLINE | ID: mdl-37377749

ABSTRACT

Implementation of ultra-high dose-rate FLASH radiotherapy (FLASH-RT) is rapidly gaining traction as a unique cancer treatment modality able to dramatically minimize normal tissue toxicity while maintaining antitumor efficacy compared with standard-of-care radiotherapy at conventional dose rate (CONV-RT). The resultant improvements in the therapeutic index have sparked intense investigations in pursuit of the underlying mechanisms. As a preamble to clinical translation, we exposed non-tumor-bearing male and female mice to hypofractionated (3 × 10 Gy) whole brain FLASH- and CONV-RT to evaluate differential neurologic responses using a comprehensive panel of functional and molecular outcomes over a 6-month follow-up. In each instance, extensive and rigorous behavioral testing showed FLASH-RT to preserve cognitive indices of learning and memory that corresponded to a similar protection of synaptic plasticity as measured by long-term potentiation (LTP). These beneficial functional outcomes were not found after CONV-RT and were linked to a preservation of synaptic integrity at the molecular (synaptophysin) level and to reductions in neuroinflammation (CD68+ microglia) throughout specific brain regions known to be engaged by our selected cognitive tasks (hippocampus, medial prefrontal cortex). Ultrastructural changes in presynaptic/postsynaptic bouton (Bassoon/Homer-1 puncta) within these same regions of the brain were not found to differ in response to dose rate. With this clinically relevant dosing regimen, we provide a mechanistic blueprint from synapse to cognition detailing how FLASH-RT reduces normal tissue complications in the irradiated brain. Significance: Functional preservation of cognition and LTP after hypofractionated FLASH-RT are linked to a protection of synaptic integrity and a reduction in neuroinflammation over protracted after irradiation times.


Subject(s)
Long-Term Potentiation , Neuroinflammatory Diseases , Male , Mice , Female , Animals , Neuronal Plasticity , Radiation Dose Hypofractionation
5.
Radiother Oncol ; 186: 109767, 2023 09.
Article in English | MEDLINE | ID: mdl-37385377

ABSTRACT

Long-term potentiation (LTP) was used to gauge the impact of conventional and FLASH dose rates on synaptic transmission. Data collected from the hippocampus and medial prefrontal cortex confirmed significant inhibition of LTP after 10 fractions of 3 Gy (30 Gy total) conventional radiotherapy. Remarkably, 10x3Gy FLASH radiotherapy and unirradiated controls were identical and exhibited normal LTP.


Subject(s)
Long-Term Potentiation , Neuronal Plasticity , Mice , Animals , Neuronal Plasticity/physiology , Long-Term Potentiation/physiology , Hippocampus/physiology , Synaptic Transmission/physiology
6.
Mol Neurodegener ; 18(1): 12, 2023 02 17.
Article in English | MEDLINE | ID: mdl-36803190

ABSTRACT

BACKGROUND: The TREM2 R47H variant is one of the strongest genetic risk factors for late-onset Alzheimer's Disease (AD). Unfortunately, many current Trem2 R47H mouse models are associated with cryptic mRNA splicing of the mutant allele that produces a confounding reduction in protein product. To overcome this issue, we developed the Trem2R47H NSS (Normal Splice Site) mouse model in which the Trem2 allele is expressed at a similar level to the wild-type Trem2 allele without evidence of cryptic splicing products. METHODS: Trem2R47H NSS mice were treated with the demyelinating agent cuprizone, or crossed with the 5xFAD mouse model of amyloidosis, to explore the impact of the TREM2 R47H variant on inflammatory responses to demyelination, plaque development, and the brain's response to plaques. RESULTS: Trem2R47H NSS mice display an appropriate inflammatory response to cuprizone challenge, and do not recapitulate the null allele in terms of impeded inflammatory responses to demyelination. Utilizing the 5xFAD mouse model, we report age- and disease-dependent changes in Trem2R47H NSS mice in response to development of AD-like pathology. At an early (4-month-old) disease stage, hemizygous 5xFAD/homozygous Trem2R47H NSS (5xFAD/Trem2R47H NSS) mice have reduced size and number of microglia that display impaired interaction with plaques compared to microglia in age-matched 5xFAD hemizygous controls. This is associated with a suppressed inflammatory response but increased dystrophic neurites and axonal damage as measured by plasma neurofilament light chain (NfL) level. Homozygosity for Trem2R47H NSS suppressed LTP deficits and loss of presynaptic puncta caused by the 5xFAD transgene array in 4-month-old mice. At a more advanced (12-month-old) disease stage 5xFAD/Trem2R47H NSS mice no longer display impaired plaque-microglia interaction or suppressed inflammatory gene expression, although NfL levels remain elevated, and a unique interferon-related gene expression signature is seen. Twelve-month old Trem2R47H NSS mice also display LTP deficits and postsynaptic loss. CONCLUSIONS: The Trem2R47H NSS mouse is a valuable model that can be used to investigate age-dependent effects of the AD-risk R47H mutation on TREM2 and microglial function including its effects on plaque development, microglial-plaque interaction, production of a unique interferon signature and associated tissue damage.


Subject(s)
Alzheimer Disease , Demyelinating Diseases , Mice , Animals , Alzheimer Disease/metabolism , Cuprizone/metabolism , RNA Splicing , Mutation , Plaque, Amyloid/pathology , Disease Models, Animal , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Microglia/metabolism , Brain/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism
7.
Cell Mol Life Sci ; 80(1): 29, 2023 Jan 06.
Article in English | MEDLINE | ID: mdl-36607431

ABSTRACT

Technological advancements have facilitated the implementation of realistic, terrestrial-based complex 33-beam galactic cosmic radiation simulations (GCR Sim) to now probe central nervous system functionality. This work expands considerably on prior, simplified GCR simulations, yielding new insights into responses of male and female mice exposed to 40-50 cGy acute or chronic radiations relevant to deep space travel. Results of the object in updated location task suggested that exposure to acute or chronic GCR Sim induced persistent impairments in hippocampus-dependent memory formation and reconsolidation in female mice that did not manifest robustly in irradiated male mice. Interestingly, irradiated male mice, but not females, were impaired in novel object recognition and chronically irradiated males exhibited increased aggressive behavior on the tube dominance test. Electrophysiology studies used to evaluate synaptic plasticity in the hippocampal CA1 region revealed significant reductions in long-term potentiation after each irradiation paradigm in both sexes. Interestingly, network-level disruptions did not translate to altered intrinsic electrophysiological properties of CA1 pyramidal cells, whereas acute exposures caused modest drops in excitatory synaptic signaling in males. Ultrastructural analyses of CA1 synapses found smaller postsynaptic densities in larger spines of chronically exposed mice compared to controls and acutely exposed mice. Myelination was also affected by GCR Sim with acutely exposed mice exhibiting an increase in the percent of myelinated axons; however, the myelin sheathes on small calibur (< 0.3 mm) and larger (> 0.5 mm) axons were thinner when compared to controls. Present findings might have been predicted based on previous studies using single and mixed beam exposures and provide further evidence that space-relevant radiation exposures disrupt critical cognitive processes and underlying neuronal network-level plasticity, albeit not to the extent that might have been previously predicted.


Subject(s)
Hippocampus , Radiation Exposure , Female , Mice , Male , Animals , Synapses , Long-Term Potentiation , Neuronal Plasticity
8.
Neuro Oncol ; 25(5): 927-939, 2023 05 04.
Article in English | MEDLINE | ID: mdl-36334265

ABSTRACT

BACKGROUND: Ultrahigh dose-rate radiotherapy (FLASH-RT) affords improvements in the therapeutic index by minimizing normal tissue toxicities without compromising antitumor efficacy compared to conventional dose-rate radiotherapy (CONV-RT). To investigate the translational potential of FLASH-RT to a human pediatric medulloblastoma brain tumor, we used a radiosensitive juvenile mouse model to assess adverse long-term neurological outcomes. METHODS: Cohorts of 3-week-old male and female C57Bl/6 mice exposed to hypofractionated (2 × 10 Gy, FLASH-RT or CONV-RT) whole brain irradiation and unirradiated controls underwent behavioral testing to ascertain cognitive status four months posttreatment. Animals were sacrificed 6 months post-irradiation and tissues were analyzed for neurological and cerebrovascular decrements. RESULTS: The neurological impact of FLASH-RT was analyzed over a 6-month follow-up. FLASH-RT ameliorated neurocognitive decrements induced by CONV-RT and preserved synaptic plasticity and integrity at the electrophysiological (long-term potentiation), molecular (synaptophysin), and structural (Bassoon/Homer-1 bouton) levels in multiple brain regions. The benefits of FLASH-RT were also linked to reduced neuroinflammation (activated microglia) and the preservation of the cerebrovascular structure, by maintaining aquaporin-4 levels and minimizing microglia colocalized to vessels. CONCLUSIONS: Hypofractionated FLASH-RT affords significant and long-term normal tissue protection in the radiosensitive juvenile mouse brain when compared to CONV-RT. The capability of FLASH-RT to preserve critical cognitive outcomes and electrophysiological properties over 6-months is noteworthy and highlights its potential for resolving long-standing complications faced by pediatric brain tumor survivors. While care must be exercised before clinical translation is realized, present findings document the marked benefits of FLASH-RT that extend from synapse to cognition and the microvasculature.


Subject(s)
Brain Neoplasms , Humans , Child , Male , Female , Animals , Mice , Disease Models, Animal , Brain Neoplasms/radiotherapy , Brain Neoplasms/etiology , Radiotherapy Dosage , Radiotherapy/adverse effects
9.
Sci Data ; 8(1): 270, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34654824

ABSTRACT

Mouse models of human diseases are invaluable tools for studying pathogenic mechanisms and testing interventions and therapeutics. For disorders such as Alzheimer's disease in which numerous models are being generated, a challenging first step is to identify the most appropriate model and age to effectively evaluate new therapeutic approaches. Here we conducted a detailed phenotypic characterization of the 5xFAD model on a congenic C57BL/6 J strain background, across its lifespan - including a seldomly analyzed 18-month old time point to provide temporally correlated phenotyping of this model and a template for characterization of new models of LOAD as they are generated. This comprehensive analysis included quantification of plaque burden, Aß biochemical levels, and neuropathology, neurophysiological measurements and behavioral and cognitive assessments, and evaluation of microglia, astrocytes, and neurons. Analysis of transcriptional changes was conducted using bulk-tissue generated RNA-seq data from microdissected cortices and hippocampi as a function of aging, which can be explored at the MODEL-AD Explorer and AD Knowledge Portal. This deep-phenotyping pipeline identified novel aspects of age-related pathology in the 5xFAD model.


Subject(s)
Alzheimer Disease/genetics , Disease Models, Animal , Phenotype , Animals , Behavior, Animal , Hippocampus , Long-Term Potentiation , Mice , Mice, Inbred C57BL , RNA-Seq , Synaptic Transmission
10.
Int J Mol Sci ; 22(16)2021 Aug 21.
Article in English | MEDLINE | ID: mdl-34445726

ABSTRACT

A recognized risk of long-duration space travel arises from the elevated exposure astronauts face from galactic cosmic radiation (GCR), which is composed of a diverse array of energetic particles. There is now abundant evidence that exposures to many different charged particle GCR components within acute time frames are sufficient to induce central nervous system deficits that span from the molecular to the whole animal behavioral scale. Enhanced spacecraft shielding can lessen exposures to charged particle GCR components, but may conversely elevate neutron radiation levels. We previously observed that space-relevant neutron radiation doses, chronically delivered at dose-rates expected during planned human exploratory missions, can disrupt hippocampal neuronal excitability, perturb network long-term potentiation and negatively impact cognitive behavior. We have now determined that acute exposures to similar low doses (18 cGy) of neutron radiation can also lead to suppressed hippocampal synaptic signaling, as well as decreased learning and memory performance in male mice. Our results demonstrate that similar nervous system hazards arise from neutron irradiation regardless of the exposure time course. While not always in an identical manner, neutron irradiation disrupts many of the same central nervous system elements as acute charged particle GCR exposures. The risks arising from neutron irradiation are therefore important to consider when determining the overall hazards astronauts will face from the space radiation environment.


Subject(s)
Cosmic Radiation/adverse effects , Hippocampus/radiation effects , Neutrons/adverse effects , Animals , Behavior, Animal/radiation effects , Male , Memory/radiation effects , Mice , Neuronal Plasticity/radiation effects
11.
Nat Commun ; 12(1): 2421, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33893290

ABSTRACT

The majority of Alzheimer's disease (AD) cases are late-onset and occur sporadically, however most mouse models of the disease harbor pathogenic mutations, rendering them better representations of familial autosomal-dominant forms of the disease. Here, we generated knock-in mice that express wildtype human Aß under control of the mouse App locus. Remarkably, changing 3 amino acids in the mouse Aß sequence to its wild-type human counterpart leads to age-dependent impairments in cognition and synaptic plasticity, brain volumetric changes, inflammatory alterations, the appearance of Periodic Acid-Schiff (PAS) granules and changes in gene expression. In addition, when exon 14 encoding the Aß sequence was flanked by loxP sites we show that Cre-mediated excision of exon 14 ablates hAß expression, rescues cognition and reduces the formation of PAS granules.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Brain/physiopathology , Disease Models, Animal , Mutation , Neuronal Plasticity/physiology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Female , Gene Expression Profiling/methods , Gene Ontology , Gene Regulatory Networks , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity/genetics
12.
Front Neurosci ; 15: 785276, 2021.
Article in English | MEDLINE | ID: mdl-35140584

ABSTRACT

Animal models of disease are valuable resources for investigating pathogenic mechanisms and potential therapeutic interventions. However, for complex disorders such as Alzheimer's disease (AD), the generation and availability of innumerous distinct animal models present unique challenges to AD researchers and hinder the success of useful therapies. Here, we conducted an in-depth analysis of the 3xTg-AD mouse model of AD across its lifespan to better inform the field of the various pathologies that appear at specific ages, and comment on drift that has occurred in the development of pathology in this line since its development 20 years ago. This modern characterization of the 3xTg-AD model includes an assessment of impairments in long-term potentiation followed by quantification of amyloid beta (Aß) plaque burden and neurofibrillary tau tangles, biochemical levels of Aß and tau protein, and neuropathological markers such as gliosis and accumulation of dystrophic neurites. We also present a novel comparison of the 3xTg-AD model with the 5xFAD model using the same deep-phenotyping characterization pipeline and show plasma NfL is strongly driven by plaque burden. The results from these analyses are freely available via the AD Knowledge Portal (https://modeladexplorer.org/). Our work demonstrates the utility of a characterization pipeline that generates robust and standardized information relevant to investigating and comparing disease etiologies of current and future models of AD.

13.
eNeuro ; 6(4)2019.
Article in English | MEDLINE | ID: mdl-31383727

ABSTRACT

As NASA prepares for a mission to Mars, concerns regarding the health risks associated with deep space radiation exposure have emerged. Until now, the impacts of such exposures have only been studied in animals after acute exposures, using dose rates ∼1.5×105 higher than those actually encountered in space. Using a new, low dose-rate neutron irradiation facility, we have uncovered that realistic, low dose-rate exposures produce serious neurocognitive complications associated with impaired neurotransmission. Chronic (6 month) low-dose (18 cGy) and dose rate (1 mGy/d) exposures of mice to a mixed field of neutrons and photons result in diminished hippocampal neuronal excitability and disrupted hippocampal and cortical long-term potentiation. Furthermore, mice displayed severe impairments in learning and memory, and the emergence of distress behaviors. Behavioral analyses showed an alarming increase in risk associated with these realistic simulations, revealing for the first time, some unexpected potential problems associated with deep space travel on all levels of neurological function.


Subject(s)
Cognition/radiation effects , Cosmic Radiation/adverse effects , Hippocampus/radiation effects , Neutrons/adverse effects , Photons/adverse effects , Synaptic Transmission/radiation effects , Animals , Anxiety/etiology , Depression/etiology , Extinction, Psychological/radiation effects , Male , Memory/radiation effects , Mice, Inbred C57BL , Neurons/radiation effects , Social Behavior
14.
Aging Cell ; 17(6): e12832, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30276955

ABSTRACT

Microglia, the resident immune cell of the brain, can be eliminated via pharmacological inhibition of the colony-stimulating factor 1 receptor (CSF1R). Withdrawal of CSF1R inhibition then stimulates microglial repopulation, effectively replacing the microglial compartment. In the aged brain, microglia take on a "primed" phenotype and studies indicate that this coincides with age-related cognitive decline. Here, we investigated the effects of replacing the aged microglial compartment with new microglia using CSF1R inhibitor-induced microglial repopulation. With 28 days of repopulation, replacement of resident microglia in aged mice (24 months) improved spatial memory and restored physical microglial tissue characteristics (cell densities and morphologies) to those found in young adult animals (4 months). However, inflammation-related gene expression was not broadly altered with repopulation nor the response to immune challenges. Instead, microglial repopulation resulted in a reversal of age-related changes in neuronal gene expression, including expression of genes associated with actin cytoskeleton remodeling and synaptogenesis. Age-related changes in hippocampal neuronal complexity were reversed with both microglial elimination and repopulation, while microglial elimination increased both neurogenesis and dendritic spine densities. These changes were accompanied by a full rescue of age-induced deficits in long-term potentiation with microglial repopulation. Thus, several key aspects of the aged brain can be reversed by acute noninvasive replacement of microglia.


Subject(s)
Aging/metabolism , Cognition/physiology , Microglia/metabolism , Neurons/metabolism , Animals , Cell Count , Cell Shape/drug effects , Cognition/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Gene Expression Regulation/drug effects , Inflammation/genetics , Inflammation/pathology , Lipopolysaccharides/pharmacology , Long-Term Potentiation/drug effects , Male , Mice, Inbred C57BL , Microglia/drug effects , Neurogenesis/drug effects , Neurons/drug effects , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Synapses/drug effects , Synapses/metabolism
15.
Nat Commun ; 9(1): 3323, 2018 08 20.
Article in English | MEDLINE | ID: mdl-30127461

ABSTRACT

Aging is accompanied by impairments in both circadian rhythmicity and long-term memory. Although it is clear that memory performance is affected by circadian cycling, it is unknown whether age-related disruption of the circadian clock causes impaired hippocampal memory. Here, we show that the repressive histone deacetylase HDAC3 restricts long-term memory, synaptic plasticity, and experience-induced expression of the circadian gene Per1 in the aging hippocampus without affecting rhythmic circadian activity patterns. We also demonstrate that hippocampal Per1 is critical for long-term memory formation. Together, our data challenge the traditional idea that alterations in the core circadian clock drive circadian-related changes in memory formation and instead argue for a more autonomous role for circadian clock gene function in hippocampal cells to gate the likelihood of long-term memory formation.


Subject(s)
Aging/physiology , Circadian Rhythm/genetics , Epigenesis, Genetic , Hippocampus/physiology , Memory/physiology , Period Circadian Proteins/genetics , Animals , Gene Deletion , Gene Knockdown Techniques , Histone Deacetylases/metabolism , Long-Term Potentiation , Memory Disorders/genetics , Memory Disorders/physiopathology , Mice, Inbred C57BL , Neuronal Plasticity/genetics , Period Circadian Proteins/metabolism
16.
Learn Mem ; 25(3): 109-114, 2018 03.
Article in English | MEDLINE | ID: mdl-29449454

ABSTRACT

Multiple epigenetic mechanisms, including histone acetylation and nucleosome remodeling, are known to be involved in long-term memory formation. Enhancing histone acetylation by deleting histone deacetylases, like HDAC3, typically enhances long-term memory formation. In contrast, disrupting nucleosome remodeling by blocking the neuron-specific chromatin remodeling subunit BAF53b impairs long-term memory. Here, we show that deleting HDAC3 can ameliorate the impairments in both long-term memory and synaptic plasticity caused by BAF53b mutation. This suggests a dynamic interplay exists between histone acetylation/deacetylation and nucleosome remodeling mechanisms in the regulation of memory formation.


Subject(s)
Histone Deacetylases/deficiency , Long-Term Potentiation/physiology , Memory Disorders/metabolism , Memory, Long-Term/physiology , Animals , Epigenesis, Genetic , Female , Hippocampus/metabolism , Histone Deacetylases/genetics , Long-Term Potentiation/genetics , Male , Memory Disorders/genetics , Mice, Inbred C57BL , Mice, Transgenic
17.
Learn Mem ; 24(5): 199-209, 2017 05.
Article in English | MEDLINE | ID: mdl-28416631

ABSTRACT

Recent human exome-sequencing studies have implicated polymorphic Brg1-associated factor (BAF) complexes (mammalian SWI/SNF chromatin remodeling complexes) in several intellectual disabilities and cognitive disorders, including autism. However, it remains unclear how mutations in BAF complexes result in impaired cognitive function. Post-mitotic neurons express a neuron-specific assembly, nBAF, characterized by the neuron-specific subunit BAF53b. Subdomain 2 of BAF53b is essential for the differentiation of neuronal precursor cells into neurons. We generated transgenic mice lacking subdomain 2 of Baf53b (BAF53bΔSB2). Long-term synaptic potentiation (LTP) and long-term memory, both of which are associated with phosphorylation of the actin severing protein cofilin, were assessed in these animals. A phosphorylation mimic of cofilin was stereotaxically delivered into the hippocampus of BAF53bΔSB2 mice in an effort to rescue LTP and memory. BAF53bΔSB2 mutant mice show impairments in phosphorylation of synaptic cofilin, LTP, and memory. Both the synaptic plasticity and memory deficits are rescued by overexpression of a phosphorylation mimetic of cofilin. Baseline physiology and behavior were not affected by the mutation or the experimental treatment. This study suggests a potential link between nBAF function, actin cytoskeletal remodeling at the dendritic spine, and memory formation. This work shows that a targeted manipulation of synaptic function can rescue adult plasticity and memory deficits caused by manipulations of nBAF, and thereby provides potential novel avenues for therapeutic development for multiple intellectual disability disorders.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/metabolism , Memory/physiology , Mutation/genetics , Neuronal Plasticity/genetics , Phosphopyruvate Hydratase/metabolism , Actin Depolymerizing Factors/genetics , Actin Depolymerizing Factors/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Nucleolus/metabolism , Chromosomal Proteins, Non-Histone/genetics , Hippocampus/cytology , Hippocampus/metabolism , In Vitro Techniques , Long-Term Potentiation/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/physiology , Neurons/ultrastructure , Phosphopyruvate Hydratase/genetics , Phosphorylation/genetics , Sequence Deletion/genetics , Transduction, Genetic
18.
Cereb Cortex ; 27(4): 2640-2651, 2017 04 01.
Article in English | MEDLINE | ID: mdl-27073215

ABSTRACT

Dendritic extension and synaptogenesis proceed at high rates in rat hippocampus during early postnatal life but markedly slow during the third week of development. The reasons for the latter, fundamental event are poorly understood. Here, we report that levels of phosphorylated (inactive) cofilin, an actin depolymerizing factor, decrease by 90% from postnatal days (pnds) 10 to 21. During the same period, levels of total and phosphorylated Arp2, which nucleates actin branches, increase. A search for elements that could explain the switch from inactive to active cofilin identified reductions in ß1 integrin, TrkB, and LIM domain kinase 2b, upstream proteins that promote cofilin phosphorylation. Moreover, levels of slingshot 3, which dephosphorylates cofilin, increase during the period in which growth slows. Consistent with the cofilin results, in situ phalloidin labeling of F-actin demonstrated that spines and dendrites contained high levels of dynamic actin filaments during Week 2, but these fell dramatically by pnd 21. The results suggest that the change from inactive to constitutively active cofilin leads to a loss of dynamic actin filaments needed for process extension and thus the termination of spine formation and synaptogenesis. The relevance of these events to the emergence of memory-related synaptic plasticity is described.


Subject(s)
Actin Depolymerizing Factors/metabolism , Hippocampus/growth & development , Hippocampus/metabolism , Neurogenesis/physiology , Neuronal Plasticity/physiology , Animals , Blotting, Western , Immunohistochemistry , Immunoprecipitation , Male , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
19.
Nat Commun ; 7: 11725, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27226355

ABSTRACT

Recent evidence implicates epigenetic mechanisms in drug-associated memory processes. However, a possible role for one major epigenetic mechanism, nucleosome remodelling, in drug-associated memories remains largely unexplored. Here we examine mice with genetic manipulations targeting a neuron-specific nucleosome remodelling complex subunit, BAF53b. These mice display deficits in cocaine-associated memory that are more severe in BAF53b transgenic mice compared with BAF53b heterozygous mice. Similar to the memory deficits, theta-induced long-term potentiation (theta-LTP) in the nucleus accumbens (NAc) is significantly impaired in slices taken from BAF53b transgenic mice but not heterozygous mice. Further experiments indicate that theta-LTP in the NAc is dependent on TrkB receptor activation, and that BDNF rescues theta-LTP and cocaine-associated memory deficits in BAF53b transgenic mice. Together, these results suggest a role for BAF53b in NAc neuronal function required for cocaine-associated memories, and also that BDNF/TrkB activation in the NAc may overcome memory and plasticity deficits linked to BAF53b mutations.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Cocaine-Related Disorders/metabolism , Memory Disorders/metabolism , Nucleus Accumbens/metabolism , Animals , Cell Line , Chromosomal Proteins, Non-Histone/genetics , Cocaine , Cocaine-Related Disorders/physiopathology , Female , Long-Term Potentiation/physiology , Male , Memory Disorders/chemically induced , Memory Disorders/physiopathology , Mice, Knockout , Mice, Transgenic , Nucleus Accumbens/physiopathology , Receptor, trkB/metabolism
20.
J Physiol ; 593(13): 2889-907, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25902928

ABSTRACT

KEY POINTS: Extended trains of theta rhythm afferent activity lead to a biphasic response facilitation in field CA1 but not in the lateral perforant path input to the dentate gyrus. Processes that reverse long-term potentiation in field CA1 are not operative in the lateral perforant path: multiple lines of evidence indicate that this reflects differences in adenosine signalling. Adenosine A1 receptors modulate baseline synaptic transmission in the lateral olfactory tract but not the associational afferents of the piriform cortex. Levels of ecto-5'-nucleotidase (CD73), an enzyme that converts extracellular ATP into adenosine, are markedly different between regions and correlate with adenosine signalling and the efficacy of theta pulse stimulation in reversing long-term potentiation. Variations in transmitter mobilization, CD73 levels, and afferent divergence result in multivariate differences in signal processing through nodes in the cortico-hippocampal network. ABSTRACT: The present study evaluated learning-related synaptic operations across the serial stages of the olfactory cortex-hippocampus network. Theta frequency stimulation produced very different time-varying responses in the Schaffer-commissural projections than in the lateral perforant path (LPP), an effect associated with distinctions in transmitter mobilization. Long-term potentiation (LTP) had a higher threshold in LPP field potential studies but not in voltage clamped neurons; coupled with input/output relationships, these results suggest that LTP threshold differences reflect the degree of input divergence. Theta pulse stimulation erased LTP in CA1 but not in the dentate gyrus (DG), although adenosine eliminated potentiation in both areas, suggesting that theta increases extracellular adenosine to a greater degree in CA1. Moreover, adenosine A1 receptor antagonism had larger effects on theta responses in CA1 than in the DG, and concentrations of ecto-5'-nucleotidase (CD73) were much higher in CA1. Input/output curves for two connections in the piriform cortex were similar to those for the LPP, whereas adenosine modulation again correlated with levels of CD73. In sum, multiple relays in a network extending from the piriform cortex through the hippocampus can be differentiated along three dimensions (input divergence, transmitter mobilization, adenosine modulation) that potently influence throughput and plasticity. A model that incorporates the regional differences, supplemented with data for three additional links, suggests that network output goes through three transitions during the processing of theta input. It is proposed that individuated relays allow the circuit to deal with different types of behavioural problems.


Subject(s)
Adenosine/metabolism , CA1 Region, Hippocampal/physiology , Long-Term Potentiation , Piriform Cortex/physiology , Synaptic Potentials , 5'-Nucleotidase/metabolism , Animals , CA1 Region, Hippocampal/metabolism , Dentate Gyrus/metabolism , Dentate Gyrus/physiology , Male , Piriform Cortex/metabolism , Rats , Rats, Sprague-Dawley , Theta Rhythm
SELECTION OF CITATIONS
SEARCH DETAIL
...