Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.518
Filter
1.
Philos Trans R Soc Lond B Biol Sci ; 379(1906): 20230223, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-38853551

ABSTRACT

Commentaries about long-term potentiation (LTP) generally proceed with an implicit assumption that largely the same physiological effect is sampled across different experiments. However, this is clearly not the case. We illustrate the point by comparing LTP in the CA3 projections to CA1 with the different forms of potentiation in the dentate gyrus. These studies lead to the hypothesis that specialized properties of CA1-LTP are adaptations for encoding unsupervised learning and episodic memory, whereas the dentate gyrus variants subserve learning that requires multiple trials and separation of overlapping bodies of information. Recent work has added sex as a second and somewhat surprising dimension along which LTP is also differentiated. Triggering events for CA1-LTP differ between the sexes and the adult induction threshold is significantly higher in females; these findings help explain why males have an advantage in spatial learning. Remarkably, the converse is true before puberty: Females have the lower LTP threshold and are better at spatial memory problems. A mechanism has been identified for the loss-of-function in females but not for the gain-of-function in males. We propose that the many and disparate demands of natural environments, with different processing requirements across ages and between sexes, led to the emergence of multiple LTPs. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.


Subject(s)
Long-Term Potentiation , Animals , Female , Humans , Male , CA1 Region, Hippocampal/physiology , CA3 Region, Hippocampal/physiology , Dentate Gyrus/physiology , Long-Term Potentiation/physiology , Memory/physiology , Sex Factors
2.
Sci Rep ; 14(1): 13168, 2024 06 07.
Article in English | MEDLINE | ID: mdl-38849397

ABSTRACT

Autism spectrum disorder (ASD) is a pervasive neurodevelopmental condition characterized by social interaction deficits, communication impairments, repetitive behaviors, and sensory sensitivities. While the etiology of ASD is multifaceted, abnormalities in glutamatergic neurotransmission and synaptic plasticity have been implicated. This study investigated the role of metabotropic glutamate receptor 8 (mGlu8) in modulating long-term potentiation (LTP) in a rat model of ASD induced by prenatal valproic acid (VPA) exposure. To induce an animal model with autism-like characteristics, pregnant rats received an intraperitoneal injection of 500 mg/kg of sodium valproate (NaVPA) on embryonic day 12.5. High-frequency stimulation was applied to the perforant path-dentate gyrus (PP-DG) synapse to induce LTP, while the mGlu8 receptor agonist (S)-3,4-dicarboxyphenylglycine (DCPG) was administered into the DG. The results revealed that VPA-exposed rats exhibited reduced LTP compared to controls. DCPG had contrasting effects, inhibiting LTP in controls and enhancing it in VPA-exposed rats. Moreover, reduced social novelty preference index (SNPI) in VPA-exposed rats was reversed by intra-DG administration of S-3,4-DCPG. In conclusion, our study advances our understanding of the complex relationship between glutamatergic neurotransmission, synaptic plasticity, and VPA-induced autism model. The findings suggest that mGlu8 receptor dysfunction plays a role in the impaired synaptic plasticity seen in ASD.


Subject(s)
Dentate Gyrus , Disease Models, Animal , Long-Term Potentiation , Prenatal Exposure Delayed Effects , Receptors, Metabotropic Glutamate , Synapses , Valproic Acid , Animals , Valproic Acid/pharmacology , Valproic Acid/adverse effects , Long-Term Potentiation/drug effects , Female , Pregnancy , Rats , Dentate Gyrus/drug effects , Synapses/drug effects , Synapses/metabolism , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/metabolism , Prenatal Exposure Delayed Effects/chemically induced , Perforant Pathway/drug effects , Autistic Disorder/chemically induced , Glycine/analogs & derivatives , Glycine/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Rats, Sprague-Dawley , Autism Spectrum Disorder/chemically induced , Male
3.
Philos Trans R Soc Lond B Biol Sci ; 379(1906): 20230221, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-38853554

ABSTRACT

Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability and is the leading known single-gene cause of autism spectrum disorder. Patients with FXS display varied behavioural deficits that include mild to severe cognitive impairments in addition to mood disorders. Currently, there is no cure for this condition; however, there is an emerging focus on therapies that inhibit mechanistic target of rapamycin (mTOR)-dependent protein synthesis owing to the clinical effectiveness of metformin for alleviating some behavioural symptoms in FXS. Adiponectin (APN) is a neurohormone that is released by adipocytes and provides an alternative means to inhibit mTOR activation in the brain. In these studies, we show that Fmr1 knockout mice, like patients with FXS, show reduced levels of circulating APN and that both long-term potentiation (LTP) and long-term depression (LTD) in the dentate gyrus (DG) are impaired. Brief (20 min) incubation of hippocampal slices in APN (50 nM) was able to rescue both LTP and LTD in the DG and increased both the surface expression and phosphorylation of GluA1 receptors. These results provide evidence for reduced APN levels in FXS playing a role in decreasing bidirectional synaptic plasticity and show that therapies which enhance APN levels may have therapeutic potential for this and related conditions.This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.


Subject(s)
Adiponectin , Dentate Gyrus , Disease Models, Animal , Fragile X Mental Retardation Protein , Fragile X Syndrome , Mice, Knockout , Neuronal Plasticity , Animals , Fragile X Syndrome/physiopathology , Fragile X Syndrome/drug therapy , Fragile X Syndrome/metabolism , Dentate Gyrus/metabolism , Dentate Gyrus/drug effects , Mice , Neuronal Plasticity/drug effects , Fragile X Mental Retardation Protein/metabolism , Fragile X Mental Retardation Protein/genetics , Adiponectin/metabolism , Long-Term Potentiation/drug effects , Male , Receptors, AMPA/metabolism
4.
Sci Rep ; 14(1): 10622, 2024 05 09.
Article in English | MEDLINE | ID: mdl-38724691

ABSTRACT

Reduced hippocampal volume occurs in major depressive disorder (MDD), potentially due to elevated glucocorticoids from an overactivated hypothalamus-pituitary-adrenal (HPA) axis. To examine this in humans, hippocampal volume and hypothalamus (HPA axis) metabolism was quantified in participants with MDD before and after antidepressant treatment. 65 participants (n = 24 males, n = 41 females) with MDD were treated in a double-blind, randomized clinical trial of escitalopram. Participants received simultaneous positron emission tomography (PET)/magnetic resonance imaging (MRI) before and after treatment. Linear mixed models examined the relationship between hippocampus/dentate gyrus volume and hypothalamus metabolism. Chi-squared tests and multivariable logistic regression examined the association between hippocampus/dentate gyrus volume change direction and hypothalamus activity change direction with treatment. Multiple linear regression compared these changes between remitter and non-remitter groups. Covariates included age, sex, and treatment type. No significant linear association was found between hippocampus/dentate gyrus volume and hypothalamus metabolism. 62% (38 of 61) of participants experienced a decrease in hypothalamus metabolism, 43% (27 of 63) of participants demonstrated an increase in hippocampus size (51% [32 of 63] for the dentate gyrus) following treatment. No significant association was found between change in hypothalamus activity and change in hippocampus/dentate gyrus volume, and this association did not vary by sex, medication, or remission status. As this multimodal study, in a cohort of participants on standardized treatment, did not find an association between hypothalamus metabolism and hippocampal volume, it supports a more complex pathway between hippocampus neurogenesis and hypothalamus metabolism changes in response to treatment.


Subject(s)
Depressive Disorder, Major , Hippocampus , Hypothalamus , Magnetic Resonance Imaging , Positron-Emission Tomography , Humans , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/diagnostic imaging , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/pathology , Male , Female , Hypothalamus/metabolism , Hypothalamus/diagnostic imaging , Adult , Hippocampus/metabolism , Hippocampus/diagnostic imaging , Hippocampus/pathology , Middle Aged , Double-Blind Method , Positron-Emission Tomography/methods , Dentate Gyrus/metabolism , Dentate Gyrus/diagnostic imaging , Dentate Gyrus/pathology , Citalopram/therapeutic use , Hypothalamo-Hypophyseal System/metabolism , Organ Size
5.
Peptides ; 178: 171244, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38788901

ABSTRACT

The neuropeptide relaxin-3 and its cognate receptor, relaxin family peptide-3 receptors (RXFP3), have been implicated in modulating learning and memory processes, but their specific roles remain unclear. This study utilized behavioral and molecular approaches to investigate the effects of putatively reversible blockade of RXFP3 in the ventral dentate gyrus (vDG) of the hippocampus on spatial and fear memory formation in rats. Male Wistar rats received bilateral vDG cannula implantation and injections of the RXFP3 antagonist, R3(BΔ23-27)R/I5 (400 ng/0.5 µL per side), or vehicle at specific time points before acquisition, consolidation, or retrieval phases of the Morris water maze and passive avoidance learning tasks. RXFP3 inhibition impaired acquisition in the passive avoidance task but not the spatial learning task. However, both memory consolidation and retrieval were disrupted in both tasks following RXFP3 antagonism. Ventral hippocampal levels of the consolidation-related kinase p70-S6 kinase (p70S6K) were reduced RXFP3 blockade. These findings highlight a key role for ventral hippocampal RXFP3 signaling in the acquisition, consolidation, and retrieval of spatial and emotional memories, extending previous work implicating this neuropeptide system in hippocampal memory processing.


Subject(s)
Dentate Gyrus , Fear , Rats, Wistar , Receptors, G-Protein-Coupled , Animals , Dentate Gyrus/metabolism , Rats , Receptors, G-Protein-Coupled/metabolism , Male , Fear/physiology , Avoidance Learning/physiology , Avoidance Learning/drug effects , Memory/physiology , Relaxin/metabolism , Spatial Memory/physiology , Spatial Memory/drug effects , Maze Learning/physiology , Maze Learning/drug effects , Hippocampus/metabolism , Hippocampus/drug effects , Receptors, Peptide/metabolism
6.
Brain Res Bull ; 213: 110975, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38734185

ABSTRACT

Chronic restraint stress induces cognitive abnormalities through changes in synapses and oxidant levels in the amygdala and hippocampus. Given the neuroprotective effects of fruit of Terminalia chebula (Halileh) in different experimental models, the present investigation aimed to address whether Terminalia chebula is able to reduce chronic restraint stress-induced behavioral, synaptic and oxidant markers in the rat model. Thirty-two male Wistar rats were randomly divided into four groups as follows: control (did not receive any treatment and were not exposed to stress), stress (restraint stress for 2 h a day for 14 consecutive days), Terminalia chebula (received 200 mg/kg hydroalcoholic extract of Terminalia chebula), and stress + Terminalia chebula groups (received 200 mg/kg extract of Terminalia chebula twenty minutes before stress) (n = 8 in each group). We used the shuttle box test to assess learning and memory, Golgi-Cox staining to examine dendritic spine density in the dentate gyrus region of the hippocampus and the basolateral and central nuclei of the amygdala, and total antioxidant capacity (TAC) and total oxidant status (TOS) in the brain. The shuttle box test results demonstrated that Terminalia chebula treatment had a profound positive effect on memory parameters, including step-through latency (STL) and time spent in the dark room, when compared to the stress group. Daily oral treatment with Terminalia chebula effectively suppressed the loss of neural spine density in the dentate gyrus region of the hippocampus and the basolateral and central nuclei of the amygdala caused by chronic restraint stress, as demonstrated by Golgi-Cox staining. Additionally, the results indicate that Terminalia chebula significantly reduced the TOS and increased TAC in the brain compared to the stress group. In conclusion, our results suggest that Terminalia chebula improved memory impairment and synaptic loss in the dentate gyrus of the hippocampus and the basolateral and central nuclei of the amygdala induced by restraint stress via inhibiting oxidative damage.


Subject(s)
Dentate Gyrus , Memory Disorders , Oxidative Stress , Plant Extracts , Rats, Wistar , Restraint, Physical , Stress, Psychological , Terminalia , Animals , Terminalia/chemistry , Male , Stress, Psychological/metabolism , Rats , Oxidative Stress/drug effects , Oxidative Stress/physiology , Dentate Gyrus/metabolism , Plant Extracts/pharmacology , Synapses/drug effects , Synapses/metabolism , Synapses/pathology , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/drug effects , Basolateral Nuclear Complex/metabolism , Basolateral Nuclear Complex/drug effects , Central Amygdaloid Nucleus/metabolism , Central Amygdaloid Nucleus/drug effects , Neuroprotective Agents/pharmacology , Dendritic Spines/drug effects , Amygdala/metabolism
7.
J Physiol Sci ; 74(1): 30, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773373

ABSTRACT

Experiments measuring evoked potentials require flexible and rapid adjustment of stimulation and recording parameters. In this study, we have developed a recording system and an associated Android application that allow making such adjustments wirelessly. The system consists of 3 units: for stimulation, recording and control. Most of the modules in this system are custom made, although the stimulator and tablet are off-the-shelf products. When installed on the tablet, our Android application allows wireless communication with the control unit from a distance of 5 m. In testing, the recording unit had low internal noise and displayed signals faithfully. Upon receiving commands from the control unit, the stimulation unit produced precisely timed pulse outputs. Using this system, we were able to record evoked field potentials in the dentate gyrus of a rat; responses increased as expected with increasing stimulation pulse amplitude and duration.


Subject(s)
Evoked Potentials , Wireless Technology , Animals , Wireless Technology/instrumentation , Rats , Evoked Potentials/physiology , Male , Electric Stimulation/methods , Dentate Gyrus/physiology
8.
Behav Brain Res ; 468: 115042, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38723676

ABSTRACT

Hippocampus is essential for episodic memory formation, lesion studies demonstrating its role especially in processing spatial and temporal information. Further, adult hippocampal neurogenesis (AHN) in the dentate gyrus (DG) has also been linked to learning. To study hippocampal neuronal activity during events like learning, in vivo calcium imaging has become increasingly popular. It relies on the use of adeno-associated viral (AAV) vectors, which seem to lead to a decrease in AHN when applied on the DG. More notably, imaging requires the implantation of a relatively large lens into the tissue. Here, we examined how injection of an AAV vector and implantation of a 1-mm-diameter lens into the dorsal DG routinely used to image calcium activity impact the behavior of adult male C57BL/6 mice. To this aim, we conducted open-field, object-recognition and object-location tasks at baseline, after AAV vector injection, and after lens implantation. Finally, we determined AHN from hippocampal slices using a doublecortin-antibody. According to our results, the operations needed for in vivo imaging of the dorsal DG did not have adverse effects on behavior, although we noticed a decrease in AHN ipsilaterally to the operations. Thus, our results suggest that in vivo imaging can be safely used to, for example, correlate patterns of calcium activity with learned behavior. One should still keep in mind that the defects on the operated side might be functionally compensated by the (hippocampus in the) contralateral hemisphere.


Subject(s)
Hippocampus , Mice, Inbred C57BL , Neurogenesis , Animals , Neurogenesis/physiology , Male , Hippocampus/metabolism , Mice , Calcium/metabolism , Behavior, Animal/physiology , Recognition, Psychology/physiology , Dentate Gyrus/metabolism , Dentate Gyrus/physiology , Dependovirus , Genetic Vectors/administration & dosage , Functional Laterality/physiology
9.
Nat Commun ; 15(1): 4047, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38744873

ABSTRACT

Human hippocampal organoids (hHOs) derived from human induced pluripotent stem cells (hiPSCs) have emerged as promising models for investigating neurodegenerative disorders, such as schizophrenia and Alzheimer's disease. However, obtaining the electrical information of these free-floating organoids in a noninvasive manner remains a challenge using commercial multi-electrode arrays (MEAs). The three-dimensional (3D) MEAs developed recently acquired only a few neural signals due to limited channel numbers. Here, we report a hippocampal cyborg organoid (cyb-organoid) platform coupling a liquid metal-polymer conductor (MPC)-based mesh neuro-interface with hHOs. The mesh MPC (mMPC) integrates 128-channel multielectrode arrays distributed on a small surface area (~2*2 mm). Stretchability (up to 500%) and flexibility of the mMPC enable its attachment to hHOs. Furthermore, we show that under Wnt3a and SHH activator induction, hHOs produce HOPX+ and PAX6+ progenitors and ZBTB20+PROX1+ dentate gyrus (DG) granule neurons. The transcriptomic signatures of hHOs reveal high similarity to the developing human hippocampus. We successfully detect neural activities from hHOs via the mMPC from this cyb-organoid. Compared with traditional planar devices, our non-invasive coupling offers an adaptor for recording neural signals from 3D models.


Subject(s)
Hippocampus , Induced Pluripotent Stem Cells , Organoids , Humans , Organoids/metabolism , Organoids/cytology , Hippocampus/cytology , Hippocampus/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Neurons/metabolism , Neurons/cytology , Metals/chemistry , Transcriptome , Dentate Gyrus/cytology , Dentate Gyrus/metabolism
10.
eNeuro ; 11(5)2024 May.
Article in English | MEDLINE | ID: mdl-38688719

ABSTRACT

Glutamatergic mossy cells (MCs) mediate associational and commissural connectivity, exhibiting significant heterogeneity along the septotemporal axis of the mouse dentate gyrus (DG). However, it remains unclear whether the neuronal features of MCs are conserved across mammals. This study compares the neuroanatomy of MCs in the DG of mice and monkeys. The MC marker, calretinin, distinguishes two subpopulations: septal and temporal. Dual-colored fluorescence labeling is utilized to compare the axonal projection patterns of these subpopulations. In both mice and monkeys, septal and temporal MCs project axons across the longitudinal axis of the ipsilateral DG, indicating conserved associational projections. However, unlike in mice, no MC subpopulations in monkeys make commissural projections to the contralateral DG. In monkeys, temporal MCs send associational fibers exclusively to the inner molecular layer, while septal MCs give rise to wide axonal projections spanning multiple molecular layers, akin to equivalent MC subpopulations in mice. Despite conserved septotemporal heterogeneity, interspecies differences are observed in the topological organization of septal MCs, particularly in the relative axonal density in each molecular layer along the septotemporal axis of the DG. In summary, this comparative analysis sheds light on both conserved and divergent features of MCs in the DG of mice and monkeys. These findings have implications for understanding functional differentiation along the septotemporal axis of the DG and contribute to our knowledge of the anatomical evolution of the DG circuit in mammals.


Subject(s)
Axons , Calbindin 2 , Dentate Gyrus , Mice, Inbred C57BL , Animals , Male , Dentate Gyrus/cytology , Dentate Gyrus/anatomy & histology , Calbindin 2/metabolism , Mossy Fibers, Hippocampal/physiology , Mice , Species Specificity , Female
11.
Neuroimage ; 292: 120607, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38614372

ABSTRACT

INTRODUCTION: In Alzheimer's disease (AD), early diagnosis facilitates treatment options and leads to beneficial outcomes for patients, their carers and the healthcare system. The neuropsychological battery of the Uniform Data Set (UDSNB3.0) assesses cognition in ageing and dementia, by measuring scores across different cognitive domains such as attention, memory, processing speed, executive function and language. However, its neuroanatomical correlates have not been investigated using 7 Tesla MRI (7T MRI). METHODS: We used 7T MRI to investigate the correlations between hippocampal subfield volumes and the UDSNB3.0 in 24 individuals with Amyloidß-status AD and 18 age-matched controls, with respective age ranges of 60 (42-76) and 62 (52-79) years. AD participants with a Medial Temporal Atrophy scale of higher than 2 on 3T MRI were excluded from the study. RESULTS: A significant difference in the entire hippocampal volume was observed in the AD group compared to healthy controls (HC), primarily influenced by CA1, the largest hippocampal subfield. Notably, no significant difference in whole brain volume between the groups implied that hippocampal volume loss was not merely reflective of overall brain atrophy. UDSNB3.0 cognitive scores showed significant differences between AD and HC, particularly in Memory, Language, and Visuospatial domains. The volume of the Dentate Gyrus (DG) showed a significant association with the Memory and Executive domain scores in AD patients as assessed by the UDSNB3.0.. The data also suggested a non-significant trend for CA1 volume associated with UDSNB3.0 Memory, Executive, and Language domain scores in AD. In a reassessment focusing on hippocampal subfields and MoCA memory subdomains in AD, associations were observed between the DG and Cued, Uncued, and Recognition Memory subscores, whereas CA1 and Tail showed associations only with Cued memory. DISCUSSION: This study reveals differences in the hippocampal volumes measured using 7T MRI, between individuals with early symptomatic AD compared with healthy controls. This highlights the potential of 7T MRI as a valuable tool for early AD diagnosis and the real-time monitoring of AD progression and treatment efficacy. CLINICALTRIALS: GOV: ID NCT04992975 (Clinicaltrial.gov 2023).


Subject(s)
Alzheimer Disease , CA1 Region, Hippocampal , Dentate Gyrus , Magnetic Resonance Imaging , Memory Disorders , Humans , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Male , Magnetic Resonance Imaging/methods , Female , Aged , Dentate Gyrus/diagnostic imaging , Dentate Gyrus/pathology , Middle Aged , CA1 Region, Hippocampal/diagnostic imaging , CA1 Region, Hippocampal/pathology , Memory Disorders/diagnostic imaging , Memory Disorders/pathology , Adult , Amyloid beta-Peptides/metabolism
12.
Brain Behav Immun ; 119: 621-636, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38670239

ABSTRACT

Rac1 is a key regulator of the cytoskeleton and neuronal plasticity, and is known to play a critical role in psychological and cognitive brain disorders. To elucidate the engram specific Rac1 signaling in fear memory, a doxycycline (Dox)-dependent robust activity marking (RAM) system was used to label dorsal dentate gyrus (DG) engram cells in mice during contextual fear conditioning. Rac1 mRNA and protein levels in DG engram cells were peaked at 24 h (day 1) after fear conditioning and were more abundant in the fear engram cells than in the non-engram cells. Optogenetic activation of Rac1 in a temporal manner in DG engram cells before memory retrieval decreased the freezing level in the fear context. Optogenetic activation of Rac1 increased autophagy protein 7 (ATG7) expression in the DG engram cells and activated DG microglia. Microglia-specific transcriptomics and fluorescence in situ hybridization revealed that overexpression of ATG7 in the fear engram cells upregulated the mRNA of Toll-like receptor TLR2/4 in DG microglia. Knockdown of microglial TLR2/4 rescued fear memory destabilization induced by ATG7 overexpression or Rac1 activation in DG engram cells. These results indicate that Rac1-driven communications between engram cells and microglia contributes to contextual fear memory destabilization, and is mediated by ATG7 and TLR2/4, and suggest a novel mechanistic framework for the cytoskeletal regulator in fear memory interference.


Subject(s)
Dentate Gyrus , Fear , Hippocampus , Memory , Microglia , Optogenetics , rac1 GTP-Binding Protein , Animals , Fear/physiology , Mice , rac1 GTP-Binding Protein/metabolism , Memory/physiology , Microglia/metabolism , Hippocampus/metabolism , Dentate Gyrus/metabolism , Male , Mice, Inbred C57BL , Autophagy-Related Protein 7/metabolism , Autophagy-Related Protein 7/genetics , Neuropeptides/metabolism , Neuronal Plasticity/physiology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/metabolism , Conditioning, Classical/physiology
13.
Sci Rep ; 14(1): 8104, 2024 04 06.
Article in English | MEDLINE | ID: mdl-38582752

ABSTRACT

GCaMP is a genetically encoded calcium indicator (GECI) widely used in neuroscience research. It measures intracellular Ca2+ level by fluorescence changes as it directly binds to Ca2+. In this process, the effect of this calcium buffer on the intracellular calcium signaling and cell physiology is often not taken into consideration. However, growing evidence from calcium imaging studies shows GCaMP expression under certain conditions can generate aberrant activity, such as seizures. In this study, we examined the effect of GCaMP6 expression in the dentate gyrus (DG) on epileptogenesis. We found that viral expression of GCaMP6s but not GCaMP6f in the DG induces tonic-clonic seizures several weeks after viral injection. Cell-type specific expression of GCaMP6s revealed the granule cells (GCs) as the key player in GCaMP6s-induced epilepsy. Finally, by using slice electrophysiology, we demonstrated that GCaMP6s expression increases neuronal excitability in the GCs. Together, this study highlights the ability of GCaMP6s in DG-associated epileptogenesis.


Subject(s)
Calcium , Neurons , Humans , Calcium/metabolism , Neurons/metabolism , Seizures/genetics , Seizures/metabolism , Calcium Signaling , Calcium, Dietary/metabolism , Dentate Gyrus/metabolism
14.
Cells ; 13(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38667284

ABSTRACT

This study investigates the combined effects of the neuropeptide Y Y1 receptor (NPY1R) agonist [Leu31-Pro34]NPY at a dose of 132 µg and Ketamine at 10 mg/Kg on cognitive functions and neuronal proliferation, against a backdrop where neurodegenerative diseases present an escalating challenge to global health systems. Utilizing male Sprague-Dawley rats in a physiological model, this research employed a single-dose administration of these compounds and assessed their impact 24 h after treatment on object-in-place memory tasks, alongside cellular proliferation within the dorsal hippocampus dentate gyrus. Methods such as the in situ proximity ligation assay and immunohistochemistry for proliferating a cell nuclear antigen (PCNA) and doublecortin (DCX) were utilized. The results demonstrated that co-administration significantly enhanced memory consolidation and increased neuronal proliferation, specifically neuroblasts, without affecting quiescent neural progenitors and astrocytes. These effects were mediated by the potential formation of NPY1R-TrkB heteroreceptor complexes, as suggested by receptor co-localization studies, although further investigation is required to conclusively prove this interaction. The findings also highlighted the pivotal role of brain-derived neurotrophic factor (BDNF) in mediating these effects. In conclusion, this study presents a promising avenue for enhancing cognitive functions and neuronal proliferation through the synergistic action of the NPY1R agonist and Ketamine, potentially via NPY1R-TrkB heteroreceptor complex formation, offering new insights into therapeutic strategies for neurodegenerative diseases.


Subject(s)
Cell Proliferation , Cognition , Doublecortin Protein , Ketamine , Neurons , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled , Receptors, Neuropeptide Y , Receptors, Neuropeptide , Animals , Male , Ketamine/pharmacology , Ketamine/administration & dosage , Cognition/drug effects , Rats , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/metabolism , Neurons/drug effects , Neurons/metabolism , Cell Proliferation/drug effects , Receptor, trkB/agonists , Receptor, trkB/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Neurogenesis/drug effects
15.
Biomolecules ; 14(4)2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38672524

ABSTRACT

Neuronal plasticity is a crucial mechanism for an adapting nervous system to change. It is shown to be regulated by perineuronal nets (PNNs), the condensed forms of the extracellular matrix (ECM) around neuronal bodies. By assessing the changes in the number, intensity, and structure of PNNs, the ultrastructure of the PNN mesh, and the expression of inhibitory and excitatory synaptic inputs on these neurons, we aimed to clarify the role of an ECM glycoprotein, tenascin-C (TnC), in the dorsal hippocampus. To enhance neuronal plasticity, TnC-deficient (TnC-/-) and wild-type (TnC+/+) young adult male mice were reared in an enriched environment (EE) for 8 weeks. Deletion of TnC in TnC-/- mice showed an ultrastructural reduction of the PNN mesh and an increased inhibitory input in the dentate gyrus (DG), and an increase in the number of PNNs with a rise in the inhibitory input in the CA2 region. EE induced an increased inhibitory input in the CA2, CA3, and DG regions; in DG, the change was also followed by an increased intensity of PNNs. No changes in PNNs or synaptic expression were found in the CA1 region. We conclude that the DG and CA2 regions emerged as focal points of alterations in PNNs and synaptogenesis with EE as mediated by TnC.


Subject(s)
Extracellular Matrix , Hippocampus , Neuronal Plasticity , Synapses , Tenascin , Animals , Tenascin/metabolism , Tenascin/genetics , Male , Mice , Hippocampus/metabolism , Extracellular Matrix/metabolism , Synapses/metabolism , Mice, Knockout , Neurons/metabolism , Mice, Inbred C57BL , Dentate Gyrus/metabolism
16.
Int J Mol Sci ; 25(8)2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38673966

ABSTRACT

Activity-dependent neuroprotective protein (ADNP) is a neuroprotective protein essential for embryonic development, proper brain development, and neuronal plasticity. Its mutation causes the autism-like ADNP syndrome (also called the Helsmoortel-Van der Aa syndrome), characterized by neural developmental disorders and motor dysfunctions. Similar to the ADNP syndrome, the ADNP haploinsufficient mouse shows low synapse density, leading to motor and cognitive ability delays. Moderate physical activity (PA) has several neuroprotective and cognitive benefits, promoting neuronal survival, differentiation, neurogenesis, and plasticity. Until now, no study has investigated the effect of moderate exercise on ADNP expression and distribution in the rat brain. The aim of the current investigation was to study the effects of moderate exercise on the ADNP expression and neuronal activation measured by the microtubule protein ß-Tubulin III. In pursuit of this objective, twenty-four rats were selected and evenly distributed into two categories: sedentary control rats and rats exposed to moderate physical activity on a treadmill over a span of 12 weeks. Our results showed that moderate PA increases the expression of ADNP and ß-Tubulin III in the dentate gyrus (DG) hippocampal region and cerebellum. Moreover, we found a co-localization of ADNP and ß-Tubulin III in both DG and cerebellum, suggesting a direct association of ADNP with adult neuronal activation induced by moderate PA.


Subject(s)
Brain , Nerve Tissue Proteins , Physical Conditioning, Animal , Animals , Male , Rats , Brain/metabolism , Cerebellum/metabolism , Dentate Gyrus/metabolism , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Neurons/metabolism , Tubulin/metabolism , Tubulin/genetics , Rats, Wistar
17.
Exp Neurol ; 377: 114801, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38685308

ABSTRACT

Anxiety and depression are the most common mental health disorders worldwide, each affecting around 30% stroke survivors. These complications not only affect the functional recovery and quality of life in stroke patients, but also are distressing for caregivers. However, effective treatments are still lacking. Enriched environment (EE), characterized with novel and multi-dimensional stimulation, has been reported to exert therapeutic effects on physical and cognitive function. In addition, EE also had potential positive effects on emotional disorders after ischemic stroke; however, the underling mechanisms have not been well elucidated. This study aimed to explore the effectiveness of EE on emotional disorders after cerebral ischemia and its underling mechanism. Sensorimotor cortical infarction was induced by photothrombosis with stable infarct location and volume, resulting in motor dysfunction, anxiety and depression-like behaviors in mice, with decreased ALFF and ReHo values and decreased c-fos expression in the infarction area and adjacent regions. Seven days' EE treatment significantly improved motor function of contralateral forelimb and exhibited anxiolytic and antidepressant effects in infarcted mice. Compared to the mice housing in a standard environment, those subjected to acute EE stimulation had significantly increased ALFF and ReHo values in the bilateral somatosensory cortex (S1, S2), dorsal dentate gyrus (dDG), dorsal CA1 of hippocampus (dCA1), lateral habenular nucleus (LHb), periaqueductal gray (PAG), ipsilateral primary motor cortex (M1), retrosplenial cortex (RSC), parietal association cortex (PtA), dorsal CA3 of hippocampus (dCA3), claustrum (Cl), ventral pallidum (VP), amygdala (Amy), and contralateral auditory cortex (Au). Some of, but not all, the ipsilateral brain regions mentioned above showed accompanying increases in c-fos expression with the most significant changes in the dDG. The number of FosB positive cells in the dDG, decreased in infarcted mice, was significantly increased after chronic EE treatment. Chemogenetic activation of dDG neurons reduced anxiety and depressive-like behaviors in infarcted mice, while neuronal inhibition resulted in void of the anxiolytic and antidepressant effects of EE. Altogether, these findings indicated that dDG neurons may mediate EE-triggered anxiolytic and antidepressant effects in cortical infarcted mice.


Subject(s)
Anxiety , Cerebral Infarction , Dentate Gyrus , Depression , Mice, Inbred C57BL , Animals , Mice , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Male , Anxiety/etiology , Anxiety/therapy , Depression/etiology , Depression/therapy , Environment , Magnetic Resonance Imaging
18.
J Biomed Inform ; 154: 104644, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38631462

ABSTRACT

OBJECTIVE: Gene expression analysis through single-cell RNA sequencing (scRNA-seq) has revolutionized our understanding of gene regulation in diverse cell types, tissues, and organisms. While existing methods primarily focus on identifying cell type-specific gene expression programs (GEPs), the characterization of GEPs associated with biological processes and stimuli responses remains limited. In this study, we aim to infer biologically meaningful GEPs that are associated with both cellular phenotypes and activity programs directly from scRNA-seq data. METHODS: We applied linear CorEx, a machine-learning-based approach, to infer GEPs by grouping genes based on total correlation optimization function in simulated and real-world scRNA-seq datasets. Additionally, we utilized a transfer learning approach to project CorEx-inferred GEPs to other scRNA-seq datasets. RESULTS: By leveraging total correlation optimization, linear CorEx groups genes and demonstrates superior performance in identifying cell types and activity programs compared to similar methods using simulated data. Furthermore, we apply this same approach to real-world scRNA-seq data from the mouse dentate gyrus and embryonic colon development, uncovering biologically relevant GEPs related to cell types, developmental ages, and cell cycle programs. We also demonstrate the potential for transfer learning by evaluating similar datasets, showcasing the cross-species sensitivity of linear CorEx. CONCLUSION: Our findings validate linear CorEx as a valuable tool for comprehensively analyzing complex signals in scRNA-seq data, leading to deeper insights into gene expression dynamics, cellular heterogeneity, and regulatory mechanisms.


Subject(s)
Machine Learning , RNA-Seq , Single-Cell Analysis , Single-Cell Analysis/methods , Animals , Mice , RNA-Seq/methods , Gene Expression Profiling/methods , Sequence Analysis, RNA/methods , Computational Biology/methods , Humans , Dentate Gyrus/metabolism , Algorithms , Colon/metabolism , Colon/cytology , Single-Cell Gene Expression Analysis
19.
Curr Biol ; 34(9): 2011-2019.e7, 2024 05 06.
Article in English | MEDLINE | ID: mdl-38636511

ABSTRACT

Environmental enrichment (EE) improves memory, particularly the ability to discriminate similar past experiences.1,2,3,4,5,6 The hippocampus supports this ability via pattern separation, the encoding of similar events using dissimilar memory representations.7 This is carried out in the dentate gyrus (DG) and CA3 subfields.8,9,10,11,12 Upregulation of adult neurogenesis in the DG improves memory through enhanced pattern separation.1,2,3,4,5,6,11,13,14,15,16 Adult-born granule cells (abGCs) in DG are suggested to contribute to pattern separation by driving inhibition in regions such as CA3,13,14,15,16,17,18 leading to sparser, nonoverlapping representations of similar events (although a role for abGCs in driving excitation in the hippocampus has also been reported16). Place cells in the hippocampus contribute to pattern separation by remapping to spatial and contextual alterations to the environment.19,20,21,22,23,24,25,26,27 How spatial responses in CA3 are affected by EE and input from increased numbers of abGCs in DG is, however, unknown. Here, we investigate the neural mechanisms facilitating improved memory following EE using associative recognition memory tasks that model the automatic and integrative nature of episodic memory. We find that EE-dependent improvements in difficult discriminations are related to increased neurogenesis and sparser memory representations across the hippocampus. Additionally, we report for the first time that EE changes how CA3 place cells discriminate similar contexts. CA3 place cells of enriched rats show greater spatial tuning, increased firing rates, and enhanced remapping to contextual changes. These findings point to more precise and flexible CA3 memory representations in enriched rats, which provides a putative mechanism for EE-dependent improvements in fine memory discrimination.


Subject(s)
CA3 Region, Hippocampal , Environment , Animals , Rats , CA3 Region, Hippocampal/physiology , Male , Neurogenesis/physiology , Rats, Long-Evans , Memory/physiology , Dentate Gyrus/physiology
20.
Expert Opin Ther Targets ; 28(4): 309-322, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38626283

ABSTRACT

BACKGROUND: Major Depressive Disorder (MDD) poses a significant challenge to global health, with current treatments often limited by efficacy and onset delays. This study explores the synergistic antidepressant-like effects of an NPY1R agonist and Ketamine, targeting their neurobiological interactions within the ventral hippocampus. RESEARCH DESIGN AND METHODS: Utilizing a preclinical model, this study administered Neuropeptide Y receptor 1 (NPY1R) agonist and Ketamine, both separately and in combination, through intracerebroventricular (icv) and intranasal (i.n.) routes. The Forced Swimming Test (FST) was employed to assess antidepressant-like activity, while in situ Proximity Ligation Assay and immunohistochemistry were used to examine NPY1R/TrkB heteroreceptor complexes and BDNF expression in the ventral dentate gyrus (DG), along with neurogenesis markers. RESULTS: The combined treatment significantly reduced immobility in the FST, indicative of enhanced antidepressant-like effects, correlated with increased formation of NPY1R/TrkB complex and brain-derived neurotrophic factor (BDNF) expression in the ventral DG. These molecular alterations were associated with increased neurogenesis. CONCLUSIONS: The coadministration of an NPY1R agonist and Ketamine in a rodent model demonstrated potentiated antidepressant responses through synergistic neurobiological pathways, including TrkB signaling and hippocampal neurogenesis. This indicates a novel therapeutic strategy for MDD, warranting further clinical investigation to fully understand its implications.


Subject(s)
Antidepressive Agents , Brain-Derived Neurotrophic Factor , Depressive Disorder, Major , Drug Synergism , Hippocampus , Ketamine , Neurogenesis , Receptor, trkB , Receptors, Neuropeptide Y , Signal Transduction , Animals , Neurogenesis/drug effects , Antidepressive Agents/pharmacology , Antidepressive Agents/administration & dosage , Male , Brain-Derived Neurotrophic Factor/metabolism , Signal Transduction/drug effects , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/metabolism , Depressive Disorder, Major/drug therapy , Ketamine/pharmacology , Ketamine/administration & dosage , Hippocampus/metabolism , Hippocampus/drug effects , Receptor, trkB/agonists , Receptor, trkB/metabolism , Disease Models, Animal , Rats , Mice , Rats, Sprague-Dawley , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Swimming
SELECTION OF CITATIONS
SEARCH DETAIL
...