Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.888
Filter
1.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article in English | MEDLINE | ID: mdl-39000499

ABSTRACT

General anesthetics may accelerate the neuropathological changes related to Alzheimer's disease (AD), of which amyloid beta (Aß)-induced toxicity is one of the main causes. However, the interaction of general anesthetics with different Aß-isoforms remains unclear. In this study, we investigated the effects of sevoflurane (0.4 and 1.2 maximal alveolar concentration (MAC)) on four Aß species-induced changes on dendritic spine density (DSD) in hippocampal brain slices of Thy1-eGFP mice and multiple epidermal growth factor-like domains 10 (MEGF10)-related astrocyte-mediated synaptic engulfment in hippocampal brain slices of C57BL/6 mice. We found that both sevoflurane and Aß downregulated CA1-dendritic spines. Moreover, compared with either sevoflurane or Aß alone, pre-treatment with Aß isoforms followed by sevoflurane application in general further enhanced spine loss. This enhancement was related to MEGF10-related astrocyte-dependent synaptic engulfment, only in AßpE3 + 1.2 MAC sevoflurane and 3NTyrAß + 1.2 MAC sevoflurane condition. In addition, removal of sevoflurane alleviated spine loss in Aß + sevoflurane. In summary, these results suggest that both synapses and astrocytes are sensitive targets for sevoflurane; in the presence of 3NTyrAß, 1.2 MAC sevoflurane alleviated astrocyte-mediated synaptic engulfment and exerted a lasting effect on dendritic spine remodeling.


Subject(s)
Amyloid beta-Peptides , Astrocytes , CA1 Region, Hippocampal , Dendritic Spines , Mice, Inbred C57BL , Sevoflurane , Synapses , Sevoflurane/pharmacology , Animals , Dendritic Spines/metabolism , Dendritic Spines/drug effects , Amyloid beta-Peptides/metabolism , Astrocytes/metabolism , Astrocytes/drug effects , Synapses/drug effects , Synapses/metabolism , Mice , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/cytology , Male , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Anesthetics, Inhalation/pharmacology
2.
J Chem Phys ; 161(3)2024 Jul 21.
Article in English | MEDLINE | ID: mdl-39007374

ABSTRACT

Voltage distribution in sub-cellular micro-domains such as neuronal synapses, small protrusions, or dendritic spines regulates the opening and closing of ionic channels, energy production, and thus, cellular homeostasis and excitability. Yet how voltage changes at such a small scale in vivo remains challenging due to the experimental diffraction limit, large signal fluctuations, and the still limited resolution of fast voltage indicators. Here, we study the voltage distribution in nano-compartments using a computational approach based on the Poisson-Nernst-Planck equations for the electro-diffusion motion of ions, where inward and outward fluxes are generated between channels. We report a current-voltage (I-V) logarithmic relationship generalizing Nernst law that reveals how the local membrane curvature modulates the voltage. We further find that an influx current penetrating a cellular electrolyte can lead to perturbations from tens to hundreds of nanometers deep, depending on the local channel organization. Finally, we show that the neck resistance of dendritic spines can be completely shunted by the transporters located on the head boundary, facilitating ionic flow. To conclude, we propose that voltage is regulated at a subcellular level by channel organization, membrane curvature, and narrow passages.


Subject(s)
Cell Membrane , Diffusion , Cell Membrane/chemistry , Cell Membrane/metabolism , Dendritic Spines/metabolism
3.
PLoS One ; 19(7): e0301063, 2024.
Article in English | MEDLINE | ID: mdl-38995900

ABSTRACT

Synaptic plasticity, the process whereby neuronal connections are either strengthened or weakened in response to stereotyped forms of stimulation, is widely believed to represent the molecular mechanism that underlies learning and memory. The holoenzyme calcium/calmodulin-dependent protein kinase II (CaMKII) plays a well-established and critical role in the induction of a variety of forms of synaptic plasticity such as long-term potentiation (LTP), long-term depression (LTD) and depotentiation. Previously, we identified the GTPase Rem2 as a potent, endogenous inhibitor of CaMKII. Here, we report that knock out of Rem2 enhances LTP at the Schaffer collateral to CA1 synapse in hippocampus, consistent with an inhibitory action of Rem2 on CaMKII in vivo. Further, re-expression of WT Rem2 rescues the enhanced LTP observed in slices obtained from Rem2 conditional knock out (cKO) mice, while expression of a mutant Rem2 construct that is unable to inhibit CaMKII in vitro fails to rescue increased LTP. In addition, we demonstrate that CaMKII and Rem2 interact in dendritic spines using a 2pFLIM-FRET approach. Taken together, our data lead us to propose that Rem2 serves as a brake on synaptic potentiation via inhibition of CaMKII activity. Further, the enhanced LTP phenotype we observe in Rem2 cKO slices reveals a previously unknown role for Rem2 in the negative regulation of CaMKII function.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Hippocampus , Long-Term Potentiation , Mice, Knockout , Synapses , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Synapses/metabolism , Synapses/physiology , Mice , Hippocampus/metabolism , Dendritic Spines/metabolism , Protein Binding
4.
Sci Rep ; 14(1): 15239, 2024 07 02.
Article in English | MEDLINE | ID: mdl-38956130

ABSTRACT

Dysbindin-1, a protein encoded by the schizophrenia susceptibility gene DTNBP1, is reduced in the hippocampus of schizophrenia patients. It is expressed in various cellular populations of the brain and implicated in dopaminergic and glutamatergic transmission. To investigate the impact of reduced dysbindin-1 in excitatory cells on hippocampal-associated behaviors and synaptic transmission, we developed a conditional knockout mouse model with deletion of dysbindin-1 gene in CaMKIIα expressing cells. We found that dysbindin-1 reduction in CaMKII expressing cells resulted in impaired spatial and social memories, and attenuation of the effects of glutamate N-methyl-d-asparate receptor (NMDAR) antagonist MK801 on locomotor activity and prepulse inhibition of startle (PPI). Dysbindin-1 deficiency in CaMKII expressing cells also resulted in reduced protein levels of NMDAR subunit GluN1 and GluN2B. These changes were associated with increased expression of immature dendritic spines in basiliar dendrites and abnormalities in excitatory synaptic transmission in the ventral hippocampus. These results highlight the functional relevance of dysbindin-1 in excitatory cells and its implication in schizophrenia-related pathologies.


Subject(s)
Dysbindin , Hippocampus , Mice, Knockout , Neurons , Receptors, N-Methyl-D-Aspartate , Synaptic Transmission , Animals , Dysbindin/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Hippocampus/metabolism , Mice , Neurons/metabolism , Schizophrenia/metabolism , Schizophrenia/pathology , Schizophrenia/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Male , Dizocilpine Maleate/pharmacology , Behavior, Animal , Dendritic Spines/metabolism , Nerve Tissue Proteins
5.
Sci Rep ; 14(1): 15855, 2024 07 09.
Article in English | MEDLINE | ID: mdl-38982271

ABSTRACT

Dendritic spines are the postsynaptic compartments of excitatory synapses, however, a substantial subset of spines additionally receives inhibitory input. In such dually innervated spines (DiSs), excitatory long-term potentiation (LTP) mechanisms are suppressed, but can be enabled by blocking tonic inhibitory GABAB receptor signaling. Here we show that LTP mechanisms at DiSs are also enabled by two other excitatory LTP stimuli. In hippocampal neurons, these chemical LTP (cLTP) stimuli induced robust movement of the Ca2+/calmodulin-dependent protein kinase II (CaMKII) to DiSs. Such synaptic CaMKII accumulation is an essential LTP mechanism at singly innervated spines (SiSs). Indeed, CaMKII accumulation at DiSs was also accompanied by other readouts for successful LTP induction: spine growth and surface insertion of GluA1. Thus, DiSs are capable of the same LTP mechanisms as SiSs, although induction of these mechanism additionally requires either reduced inhibitory signaling or increased excitatory stimulation. This additional regulation may provide further computational control.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Dendritic Spines , Long-Term Potentiation , Dendritic Spines/metabolism , Dendritic Spines/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Hippocampus/metabolism , Hippocampus/cytology , Hippocampus/physiology , Synapses/physiology , Synapses/metabolism , Receptors, AMPA/metabolism , Rats , Neurons/metabolism , Neurons/physiology
6.
Mol Brain ; 17(1): 33, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38840181

ABSTRACT

Loss-of-function mutations in the progranulin (GRN) gene are an autosomal dominant cause of Frontotemporal Dementia (FTD). These mutations typically result in haploinsufficiency of the progranulin protein. Grn+/- mice provide a model for progranulin haploinsufficiency and develop FTD-like behavioral abnormalities by 9-10 months of age. In previous work, we demonstrated that Grn+/- mice develop a low dominance phenotype in the tube test that is associated with reduced dendritic arborization of layer II/III pyramidal neurons in the prelimbic region of the medial prefrontal cortex (mPFC), a region key for social dominance behavior in the tube test assay. In this study, we investigated whether progranulin haploinsufficiency induced changes in dendritic spine density and morphology. Individual layer II/III pyramidal neurons in the prelimbic mPFC of 9-10 month old wild-type or Grn+/- mice were targeted for iontophoretic microinjection of fluorescent dye, followed by high-resolution confocal microscopy and 3D reconstruction for morphometry analysis. Dendritic spine density in Grn+/- mice was comparable to wild-type littermates, but the apical dendrites in Grn+/- mice had a shift in the proportion of spine types, with fewer stubby spines and more thin spines. Additionally, apical dendrites of Grn+/- mice had longer spines and smaller thin spine head diameter in comparison to wild-type littermates. These changes in spine morphology may contribute to altered circuit-level activity and social dominance deficits in Grn+/- mice.


Subject(s)
Dendritic Spines , Haploinsufficiency , Prefrontal Cortex , Progranulins , Animals , Dendritic Spines/metabolism , Prefrontal Cortex/pathology , Prefrontal Cortex/metabolism , Progranulins/deficiency , Progranulins/genetics , Mice , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Male , Mice, Inbred C57BL
7.
Eur J Pharmacol ; 977: 176756, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38897021

ABSTRACT

Repeated exposure to propofol during early brain development is associated with anxiety disorders in adulthood, yet the mechanisms underlying propofol-induced susceptibility to anxiety disorders remain elusive. The lateral septum (LS), primarily composed of γ-aminobutyric acidergic (GABAergic) neurons, serves as a key brain region in the regulation of anxiety. However, it remains unclear whether LS GABAergic neurons are implicated in propofol-induced anxiety. Therefore, we conducted c-Fos immunostaining of whole-brain slices from mice exposed to propofol during early life. Our findings indicate that propofol exposure activates GABAergic neurons in the LS. Selective activation of LS GABAergic neurons resulted in increased anxiety-like behavior, while selective inhibition of these neurons reduced such behaviors. These results suggest that the LS is a critical brain region involved in propofol-induced anxiety. Furthermore, we investigated the molecular mechanism of propofol-induced anxiety in the LS. Microglia activation underlies the development of anxiety. Immunofluorescence staining and Western blot analysis of LS revealed activated microglia and significantly elevated levels of phospho-NF-κB p65 protein. Additionally, a decrease in the number of neuronal spines was observed. Our study highlights the crucial role of the LS in the development of anxiety-like behavior in adulthood following childhood propofol exposure, accompanied by the activation of inflammatory pathways.


Subject(s)
Anxiety , Behavior, Animal , GABAergic Neurons , Microglia , Propofol , Propofol/pharmacology , Animals , Anxiety/chemically induced , Mice , Male , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Behavior, Animal/drug effects , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Proto-Oncogene Proteins c-fos/metabolism , Mice, Inbred C57BL , Transcription Factor RelA/metabolism , Dendritic Spines/drug effects , Dendritic Spines/pathology , Dendritic Spines/metabolism
8.
J Neurosci ; 44(28)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38830758

ABSTRACT

Shank3 is a synaptic scaffolding protein that assists in tethering and organizing structural proteins and glutamatergic receptors in the postsynaptic density of excitatory synapses. The localization of Shank3 at excitatory synapses and the formation of stable Shank3 complexes is regulated by the binding of zinc to the C-terminal sterile-alpha-motif (SAM) domain of Shank3. Mutations in the SAM domain of Shank3 result in altered synaptic function and morphology, and disruption of zinc in synapses that express Shank3 leads to a reduction of postsynaptic proteins important for synaptic structure and function. This suggests that zinc supports the localization of postsynaptic proteins via Shank3. Many regions of the brain are highly enriched with free zinc inside glutamatergic vesicles at presynaptic terminals. At these synapses, zinc transporter 3 (ZnT3) moves zinc into vesicles where it is co-released with glutamate. Alterations in ZnT3 are implicated in multiple neurodevelopmental disorders, and ZnT3 knock-out (KO) mice-which lack synaptic zinc-show behavioral deficits associated with autism spectrum disorder and schizophrenia. Here we show that male and female ZnT3 KO mice have smaller dendritic spines and miniature excitatory postsynaptic current amplitudes than wildtype (WT) mice in the auditory cortex. Additionally, spine size deficits in ZnT3 KO mice are restricted to synapses that express Shank3. In WT mice, synapses that express both Shank3 and ZnT3 have larger spines compared to synapses that express Shank3 but not ZnT3. Together these findings suggest a mechanism whereby presynaptic ZnT3-dependent zinc supports postsynaptic structure and function via Shank3 in a synapse-specific manner.


Subject(s)
Auditory Cortex , Cation Transport Proteins , Dendritic Spines , Nerve Tissue Proteins , Synapses , Animals , Mice , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Synapses/metabolism , Dendritic Spines/metabolism , Cation Transport Proteins/metabolism , Cation Transport Proteins/genetics , Auditory Cortex/metabolism , Female , Male , Mice, Knockout , Carrier Proteins/metabolism , Carrier Proteins/genetics , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Excitatory Postsynaptic Potentials/physiology
9.
J Neurosci ; 44(28)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38858079

ABSTRACT

Tau pathologies are detected in the brains of some of the most common neurodegenerative diseases including Alzheimer's disease (AD), Lewy body dementia (LBD), chronic traumatic encephalopathy (CTE), and frontotemporal dementia (FTD). Tau proteins are expressed in six isoforms with either three or four microtubule-binding repeats (3R tau or 4R tau) due to alternative RNA splicing. AD, LBD, and CTE brains contain pathological deposits of both 3R and 4R tau. FTD patients can exhibit either 4R tau pathologies in most cases or 3R tau pathologies less commonly in Pick's disease, which is a subfamily of FTD. Here, we report the isoform-specific roles of tau in FTD. The P301L mutation, linked to familial 4R tau FTD, induces mislocalization of 4R tau to dendritic spines in primary hippocampal cultures that were prepared from neonatal rat pups of both sexes. Contrastingly, the G272V mutation, linked to familial Pick's disease, induces phosphorylation-dependent mislocalization of 3R tau but not 4R tau proteins to dendritic spines. The overexpression of G272V 3R tau but not 4R tau proteins leads to the reduction of dendritic spine density and suppression of mEPSCs in 5-week-old primary rat hippocampal cultures. The decrease in mEPSC amplitude caused by G272V 3R tau is dynamin-dependent whereas that caused by P301L 4R tau is dynamin-independent, indicating that the two tau isoforms activate different signaling pathways responsible for excitatory synaptic dysfunction. Our 3R and 4R tau studies here will shed new light on diverse mechanisms underlying FTD, AD, LBD, and CTE.


Subject(s)
Dendritic Spines , Frontotemporal Dementia , Mutation , Protein Isoforms , tau Proteins , tau Proteins/metabolism , tau Proteins/genetics , Animals , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Dendritic Spines/metabolism , Dendritic Spines/pathology , Rats , Male , Humans , Female , Protein Isoforms/genetics , Protein Isoforms/metabolism , Synapses/metabolism , Synapses/pathology , Rats, Sprague-Dawley , Hippocampus/metabolism , Hippocampus/pathology , Cells, Cultured
10.
J Comp Neurol ; 532(7): e25645, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38943486

ABSTRACT

Dendritic spines are sites of synaptic plasticity and their head size correlates with the strength of the corresponding synapse. We recently showed that the distribution of spine head sizes follows a lognormal-like distribution even after blockage of activity or plasticity induction. As the cytokine tumor necrosis factor (TNF) influences synaptic transmission and constitutive TNF and receptor (TNF-R)-deficiencies cause changes in spine head size distributions, we tested whether these genetic alterations disrupt the lognormality of spine head sizes. Furthermore, we distinguished between spines containing the actin-modulating protein synaptopodin (SP-positive), which is present in large, strong and stable spines and those lacking it (SP-negative). Our analysis revealed that neither TNF-deficiency nor the absence of TNF-R1, TNF-R2 or TNF-R 1 and 2 (TNF-R1/R2) degrades the general lognormal-like, skewed distribution of spine head sizes (all spines, SP-positive spines, SP-negative spines). However, TNF, TNF-R1 and TNF-R2-deficiency affected the width of the lognormal distribution, and TNF-R1/2-deficiency shifted the distribution to the left. Our findings demonstrate the robustness of the lognormal-like, skewed distribution, which is maintained even in the face of genetic manipulations that alter the distribution of spine head sizes. Our observations are in line with homeostatic adaptation mechanisms of neurons regulating the distribution of spines and their head sizes.


Subject(s)
Dendritic Spines , Dentate Gyrus , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor, Type II , Receptors, Tumor Necrosis Factor, Type I , Tumor Necrosis Factor-alpha , Animals , Dendritic Spines/metabolism , Mice , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Dentate Gyrus/metabolism , Dentate Gyrus/cytology , Tumor Necrosis Factor-alpha/metabolism , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type II/metabolism , Receptors, Tumor Necrosis Factor, Type II/genetics , Neurons/metabolism , Male , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/deficiency
11.
J Neurosci ; 44(29)2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38886056

ABSTRACT

The small G-protein Ras-related C3 botulinum toxin substrate 1 (Rac1) promotes the formation of filamentous actin (F-actin). Actin is a major component of dendritic spines, and we previously found that alcohol alters actin composition and dendritic spine structure in the nucleus accumbens (NAc) and the dorsomedial striatum (DMS). To examine if Rac1 contributes to these alcohol-mediated adaptations, we measured the level of GTP-bound active Rac1 in the striatum of mice following 7 weeks of intermittent access to 20% alcohol. We found that chronic alcohol intake activates Rac1 in the DMS of male mice. In contrast, Rac1 is not activated by alcohol in the NAc and DLS of male mice or in the DMS of female mice. Similarly, closely related small G-proteins are not activated by alcohol in the DMS, and Rac1 activity is not increased in the DMS by moderate alcohol or natural reward. To determine the consequences of alcohol-dependent Rac1 activation in the DMS of male mice, we inhibited endogenous Rac1 by infecting the DMS of mice with an adeno-associated virus (AAV) expressing a dominant negative form of the small G-protein (Rac1-DN). We found that overexpression of AAV-Rac1-DN in the DMS inhibits alcohol-mediated Rac1 signaling and attenuates alcohol-mediated F-actin polymerization, which corresponded with a decrease in dendritic arborization and spine maturation. Finally, we provide evidence to suggest that Rac1 in the DMS plays a role in alcohol-associated goal-directed learning. Together, our data suggest that Rac1 in the DMS plays an important role in alcohol-dependent structural plasticity and aberrant learning.


Subject(s)
Corpus Striatum , Mice, Inbred C57BL , Neuronal Plasticity , rac1 GTP-Binding Protein , Animals , Male , Mice , rac1 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/genetics , Neuronal Plasticity/physiology , Neuronal Plasticity/drug effects , Female , Corpus Striatum/metabolism , Corpus Striatum/drug effects , Ethanol/pharmacology , Learning/physiology , Learning/drug effects , Neuropeptides/metabolism , Neuropeptides/genetics , Dendritic Spines/metabolism , Dendritic Spines/drug effects
12.
Behav Brain Res ; 471: 115097, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38878971

ABSTRACT

Neuroadaptive changes in the hippocampus underlie addictive-like behaviors in humans or animals chronically exposed to cocaine. miR-181a, which is widely expressed in the hippocampus, acts as a regulator for synaptic plasticity, while its role in drug reinstatement is unclear. In this study, we found that miR-181a regulates the reinstatement of cocaine conditioned place preference(CPP), and altered miR-181a expression changes the complexity of hippocampal neurons and the density and morphology of dendritic spines. By using a luciferase gene reporter, we found that miR-181a targets PRKAA1, an upstream molecule in the mTOR pathway. High miR-181a expression reduced the expression of the PRKAA1 mRNA and promoted mTOR activity and the reinstatement of cocaine CPP. These results indicate that miR-181a is involved in neuronal structural plasticity induced by reinstatement of cocaine CPP, possibly through the activation of the mTOR signaling pathway. This study provides new microRNA targets and a theoretical foundation for the prevention of cocaine-induced reinstatement.


Subject(s)
Cocaine , Hippocampus , MicroRNAs , TOR Serine-Threonine Kinases , MicroRNAs/metabolism , Animals , Hippocampus/metabolism , Hippocampus/drug effects , Cocaine/pharmacology , Male , TOR Serine-Threonine Kinases/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , AMP-Activated Protein Kinases/metabolism , Neurons/drug effects , Neurons/metabolism , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Dopamine Uptake Inhibitors/pharmacology , Cocaine-Related Disorders/metabolism , Mice , Rats , Rats, Sprague-Dawley
13.
Cells ; 13(11)2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38891120

ABSTRACT

Methyl-CpG-binding protein 2 (Mecp2) is an epigenetic modulator and numerous studies have explored its impact on the central nervous system manifestations. However, little attention has been given to its potential contributions to the peripheral nervous system (PNS). To investigate the regulation of Mecp2 in the PNS on specific central regions, we generated Mecp2fl/flAdvillincre mice with the sensory-neuron-specific deletion of the Mecp2 gene and found the mutant mice had a heightened sensitivity to temperature, which, however, did not affect the sense of motion, social behaviors, and anxiety-like behavior. Notably, in comparison to Mecp2fl/fl mice, Mecp2fl/flAdvillincre mice exhibited improved learning and memory abilities. The levels of hippocampal synaptophysin and PSD95 proteins were higher in Mecp2fl/flAdvillincre mice than in Mecp2fl/fl mice. Golgi staining revealed a significant increase in total spine density, and dendritic arborization in the hippocampal pyramidal neurons of Mecp2fl/flAdvillincre mice compared to Mecp2fl/fl mice. In addition, the activation of the BDNF-TrkB-CREB1 pathway was observed in the hippocampus and spinal cord of Mecp2fl/flAdvillincre mice. Intriguingly, the hippocampal BDNF/CREB1 signaling pathway in mutant mice was initiated within 5 days after birth. Our findings suggest a potential therapeutic strategy targeting the BDNF-TrkB-CREB1 signaling pathway and peripheral somasensory neurons to treat learning and cognitive deficits associated with Mecp2 disorders.


Subject(s)
Brain-Derived Neurotrophic Factor , Cognition , Dendritic Spines , Hippocampus , Methyl-CpG-Binding Protein 2 , Animals , Methyl-CpG-Binding Protein 2/metabolism , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/deficiency , Hippocampus/metabolism , Hippocampus/pathology , Dendritic Spines/metabolism , Mice , Brain-Derived Neurotrophic Factor/metabolism , Sensory Receptor Cells/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Male , Signal Transduction , Mice, Inbred C57BL , Receptor, trkB/metabolism , Receptor, trkB/genetics
14.
Zool Res ; 45(3): 535-550, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38747058

ABSTRACT

Proper regulation of synapse formation and elimination is critical for establishing mature neuronal circuits and maintaining brain function. Synaptic abnormalities, such as defects in the density and morphology of postsynaptic dendritic spines, underlie the pathology of various neuropsychiatric disorders. Protocadherin 17 (PCDH17) is associated with major mood disorders, including bipolar disorder and depression. However, the molecular mechanisms by which PCDH17 regulates spine number, morphology, and behavior remain elusive. In this study, we found that PCDH17 functions at postsynaptic sites, restricting the number and size of dendritic spines in excitatory neurons. Selective overexpression of PCDH17 in the ventral hippocampal CA1 results in spine loss and anxiety- and depression-like behaviors in mice. Mechanistically, PCDH17 interacts with actin-relevant proteins and regulates actin filament (F-actin) organization. Specifically, PCDH17 binds to ROCK2, increasing its expression and subsequently enhancing the activity of downstream targets such as LIMK1 and the phosphorylation of cofilin serine-3 (Ser3). Inhibition of ROCK2 activity with belumosudil (KD025) ameliorates the defective F-actin organization and spine structure induced by PCDH17 overexpression, suggesting that ROCK2 mediates the effects of PCDH17 on F-actin content and spine development. Hence, these findings reveal a novel mechanism by which PCDH17 regulates synapse development and behavior, providing pathological insights into the neurobiological basis of mood disorders.


Subject(s)
Actin Cytoskeleton , Cadherins , Dendritic Spines , Protocadherins , rho-Associated Kinases , Animals , Mice , Actin Cytoskeleton/metabolism , Cadherins/metabolism , Cadherins/genetics , Dendritic Spines/metabolism , Dendritic Spines/physiology , Gene Expression Regulation , rho-Associated Kinases/metabolism , rho-Associated Kinases/genetics , Protocadherins/genetics , Protocadherins/metabolism
15.
Int J Mol Sci ; 25(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38791517

ABSTRACT

Maternal immune activation (MIA) is a risk factor for multiple neurodevelopmental disorders; however, animal models developed to explore MIA mechanisms are sensitive to experimental factors, which has led to complexity in previous reports of the MIA phenotype. We sought to characterize an MIA protocol throughout development to understand how prenatal immune insult alters the trajectory of important neurodevelopmental processes, including the microglial regulation of synaptic spines and complement signaling. We used polyinosinic:polycytidylic acid (polyI:C) to induce MIA on gestational day 9.5 in CD-1 mice, and measured their synaptic spine density, microglial synaptic pruning, and complement protein expression. We found reduced dendritic spine density in the somatosensory cortex starting at 3-weeks-of-age with requisite increases in microglial synaptic pruning and phagocytosis, suggesting spine density loss was caused by increased microglial synaptic pruning. Additionally, we showed dysregulation in complement protein expression persisting into adulthood. Our findings highlight disruptions in the prenatal environment leading to alterations in multiple dynamic processes through to postnatal development. This could potentially suggest developmental time points during which synaptic processes could be measured as risk factors or targeted with therapeutics for neurodevelopmental disorders.


Subject(s)
Complement System Proteins , Dendritic Spines , Microglia , Poly I-C , Animals , Microglia/metabolism , Microglia/drug effects , Microglia/immunology , Mice , Female , Pregnancy , Dendritic Spines/metabolism , Poly I-C/pharmacology , Complement System Proteins/metabolism , Complement System Proteins/immunology , Prenatal Exposure Delayed Effects , Phagocytosis , Disease Models, Animal , Somatosensory Cortex/drug effects , Somatosensory Cortex/metabolism , Synapses/metabolism , Synapses/drug effects , Neuronal Plasticity/drug effects
16.
Sci Rep ; 14(1): 12252, 2024 05 28.
Article in English | MEDLINE | ID: mdl-38806649

ABSTRACT

Sex hormones affect structural and functional plasticity in the rodent hippocampus. However, hormone levels not only differ between males and females, but also fluctuate across the female estrous cycle. While sex- and cycle-dependent differences in dendritic spine density and morphology have been found in the rodent CA1 region, but not in the CA3 or the dentate gyrus, comparable structural data on CA2, i.e. the hippocampal region involved in social recognition memory, is so far lacking. In this study, we, therefore, used wildtype male and female mice in diestrus or proestrus to analyze spines on dendritic segments from identified CA2 neurons. In basal stratum oriens, we found no differences in spine density, but a significant shift towards larger spine head areas in male mice compared to females. Conversely, in apical stratum radiatum diestrus females had a significantly higher spine density, and females in either cycle stage had a significant shift towards larger spine head areas as compared to males, with diestrus females showing the larger shift. Our results provide further evidence for the sexual dimorphism of hippocampal area CA2, and underscore the importance of considering not only the sex, but also the stage of the estrous cycle when interpreting morphological data.


Subject(s)
CA2 Region, Hippocampal , Dendritic Spines , Estrous Cycle , Animals , Male , Female , Dendritic Spines/metabolism , Dendritic Spines/physiology , Mice , Estrous Cycle/physiology , CA2 Region, Hippocampal/physiology , CA2 Region, Hippocampal/metabolism , Sex Characteristics , Neurons/metabolism
17.
Sci Rep ; 14(1): 11713, 2024 05 22.
Article in English | MEDLINE | ID: mdl-38778177

ABSTRACT

The development of neurons is regulated by several spatiotemporally changing factors, which are crucial to give the ability of neurons to form functional networks. While external physical stimuli may impact the early developmental stages of neurons, the medium and long-term consequences of these influences have yet to be thoroughly examined. Using an animal model, this study focuses on the morphological and transcriptome changes of the hippocampus that may occur as a consequence of fetal ultrasound examination. We selectively labeled CA1 neurons of the hippocampus with in-utero electroporation to analyze their morphological features. Furthermore, certain samples also went through RNA sequencing after repetitive ultrasound exposure. US exposure significantly changed several morphological properties of the basal dendritic tree. A notable increase was also observed in the density of spines on the basal dendrites, accompanied by various alterations in individual spine morphology. Transcriptome analysis revealed several up or downregulated genes, which may explain the molecular background of these alterations. Our results suggest that US-derived changes in the dendritic trees of CA1 pyramidal cells might be connected to modification of the transcriptome of the hippocampus and may lead to an increased dendritic input.


Subject(s)
CA1 Region, Hippocampal , Dendrites , Transcriptome , Animals , CA1 Region, Hippocampal/metabolism , Dendrites/metabolism , Female , Pregnancy , Pyramidal Cells/metabolism , Mice , Hippocampus/metabolism , Gene Expression Profiling , Dendritic Spines/metabolism , Ultrasonography, Prenatal
18.
Neuropharmacology ; 254: 109988, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38744401

ABSTRACT

Neuropathic pain (NP) is usually treated with analgesics and symptomatic therapy with poor efficacy and numerous side effects, highlighting the urgent need for effective treatment strategies. Recent studies have reported an important role for peroxisome proliferator-activated receptor alpha (PPARα) in regulating metabolism as well as inflammatory responses. Through pain behavioral assessment, we found that activation of PPARα prevented chronic constriction injury (CCI)-induced mechanical allodynia and thermal hyperalgesia. In addition, PPARα ameliorated inflammatory cell infiltration at the injury site and decreased microglial activation, NOD-like receptor protein 3 (NLRP3) inflammasome production, and spinal dendritic spine density, as well as improved serum and spinal cord metabolic levels in mice. Administration of PPARα antagonists eliminates the analgesic effect of PPARα agonists. PPARα relieves NP by inhibiting neuroinflammation and functional synaptic plasticity as well as modulating metabolic mechanisms, suggesting that PPARα may be a potential molecular target for NP alleviation. However, the effects of PPARα on neuroinflammation and synaptic plasticity should be further explored.


Subject(s)
Mice, Inbred C57BL , Neuralgia , PPAR alpha , Spinal Cord , Animals , PPAR alpha/metabolism , Neuralgia/drug therapy , Neuralgia/metabolism , Male , Mice , Spinal Cord/metabolism , Spinal Cord/drug effects , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Metabolomics , Microglia/drug effects , Microglia/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Dendritic Spines/pathology , Inflammasomes/metabolism , Inflammasomes/drug effects
19.
Commun Biol ; 7(1): 642, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802535

ABSTRACT

Alterations in the experience-dependent and autonomous elaboration of neural circuits are assumed to underlie autism spectrum disorder (ASD), though it is unclear what synaptic traits are responsible. Here, utilizing a valproic acid-induced ASD marmoset model, which shares common molecular features with idiopathic ASD, we investigate changes in the structural dynamics of tuft dendrites of upper-layer pyramidal neurons and adjacent axons in the dorsomedial prefrontal cortex through two-photon microscopy. In model marmosets, dendritic spine turnover is upregulated, and spines are generated in clusters and survived more often than in control marmosets. Presynaptic boutons in local axons, but not in commissural long-range axons, demonstrate hyperdynamic turnover in model marmosets, suggesting alterations in projection-specific plasticity. Intriguingly, nasal oxytocin administration attenuates clustered spine emergence in model marmosets. Enhanced clustered spine generation, possibly unique to certain presynaptic partners, may be associated with ASD and be a potential therapeutic target.


Subject(s)
Callithrix , Disease Models, Animal , Neuronal Plasticity , Oxytocin , Animals , Oxytocin/metabolism , Male , Synapses/metabolism , Dendritic Spines/metabolism , Dendritic Spines/pathology , Dendritic Spines/drug effects , Autism Spectrum Disorder/metabolism , Autistic Disorder/metabolism , Autistic Disorder/pathology , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Prefrontal Cortex/drug effects , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Valproic Acid/pharmacology , Presynaptic Terminals/metabolism , Female , Axons/metabolism
20.
Dis Model Mech ; 17(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38785269

ABSTRACT

Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in MECP2, which encodes methyl-CpG-binding protein 2, a transcriptional regulator of many genes, including brain-derived neurotrophic factor (BDNF). BDNF levels are lower in multiple brain regions of Mecp2-deficient mice, and experimentally increasing BDNF levels improve atypical phenotypes in Mecp2 mutant mice. Due to the low blood-brain barrier permeability of BDNF itself, we tested the effects of LM22A-4, a brain-penetrant, small-molecule ligand of the BDNF receptor TrkB (encoded by Ntrk2), on dendritic spine density and form in hippocampal pyramidal neurons and on behavioral phenotypes in female Mecp2 heterozygous (HET) mice. A 4-week systemic treatment of Mecp2 HET mice with LM22A-4 restored spine volume in MeCP2-expressing neurons to wild-type (WT) levels, whereas spine volume in MeCP2-lacking neurons remained comparable to that in neurons from female WT mice. Female Mecp2 HET mice engaged in aggressive behaviors more than WT mice, the levels of which were reduced to WT levels by the 4-week LM22A-4 treatment. These data provide additional support to the potential usefulness of novel therapies not only for RTT but also to other BDNF-related disorders.


Subject(s)
Behavior, Animal , Dendritic Spines , Methyl-CpG-Binding Protein 2 , Phenotype , Receptor, trkB , Rett Syndrome , Animals , Rett Syndrome/pathology , Rett Syndrome/drug therapy , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Dendritic Spines/pathology , Female , Receptor, trkB/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Methyl-CpG-Binding Protein 2/genetics , Behavior, Animal/drug effects , Ligands , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Mice , Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/pathology , Hippocampus/metabolism , Hippocampus/drug effects , Heterozygote , Mice, Inbred C57BL , Disease Models, Animal , Benzamides
SELECTION OF CITATIONS
SEARCH DETAIL