Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122.685
Filter
1.
Neurosignals ; 31(1): 1-25, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38967556

ABSTRACT

Hallucination is a sensory perception that occurs in the absence of external stimuli during abnormal neurological disturbances and various mental diseases. Hallucination is recognized as a core psychotic symptom and is particularly more prevalent in individuals with schizophrenia. Strikingly, a significant number of subjects with Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and other neurological diseases like cerebral stroke and epileptic seizure also experience hallucination. While aberrant neurotransmission has been linked to the neuropathogenic events of schizophrenia, the precise cellular mechanism accounting for hallucinations remains obscure. Neurogenesis is a cellular process of producing new neurons from the neural stem cells (NSC)-derived neuroblasts in the brain that contribute to the regulation of pattern separation, mood, olfaction, learning, and memory in adulthood. Impaired neurogenesis in the hippocampus of the adult brain has been linked to stress, anxiety, depression, and dementia. Notably, many neurodegenerative disorders are characterized by the mitotic and functional activation of neuroblasts and cell cycle re-entry of mature neurons leading to a drastic alteration in neurogenic process, known as reactive neuroblastosis. Considering their neurophysiological properties, the abnormal integration of neuroblasts into the existing neural network or withdrawal of their connections can lead to abnormal synaptogenesis, and neurotransmission. Eventually, this would be expected to result in altered perception accounting for hallucination. Thus, this article emphasizes a hypothesis that aberrant neurogenic processes at the level of reactive neuroblastosis could be an underlying mechanism of hallucination in schizophrenia and other neurological diseases.


Subject(s)
Hallucinations , Hippocampus , Neurogenesis , Neuronal Plasticity , Schizophrenia , Humans , Schizophrenia/pathology , Schizophrenia/physiopathology , Hallucinations/pathology , Hallucinations/physiopathology , Neuronal Plasticity/physiology , Hippocampus/pathology , Neurogenesis/physiology , Animals , Neural Stem Cells/pathology , Neurons/pathology , Neurons/metabolism
2.
Sci Rep ; 14(1): 15338, 2024 07 03.
Article in English | MEDLINE | ID: mdl-38961135

ABSTRACT

Blood-brain barrier (BBB) disruption may contribute to cognitive decline, but questions remain whether this association is more pronounced for certain brain regions, such as the hippocampus, or represents a whole-brain mechanism. Further, whether human BBB leakage is triggered by excessive vascular pulsatility, as suggested by animal studies, remains unknown. In a prospective cohort (N = 50; 68-84 years), we used contrast-enhanced MRI to estimate the permeability-surface area product (PS) and fractional plasma volume ( v p ), and 4D flow MRI to assess cerebral arterial pulsatility. Cognition was assessed by the Montreal Cognitive Assessment (MoCA) score. We hypothesized that high PS would be associated with high arterial pulsatility, and that links to cognition would be specific to hippocampal PS. For 15 brain regions, PS ranged from 0.38 to 0.85 (·10-3 min-1) and v p from 0.79 to 1.78%. Cognition was related to PS (·10-3 min-1) in hippocampus (ß = - 2.9; p = 0.006), basal ganglia (ß = - 2.3; p = 0.04), white matter (ß = - 2.6; p = 0.04), whole-brain (ß = - 2.7; p = 0.04) and borderline-related for cortex (ß = - 2.7; p = 0.076). Pulsatility was unrelated to PS for all regions (p > 0.19). Our findings suggest PS-cognition links mainly reflect a whole-brain phenomenon with only slightly more pronounced links for the hippocampus, and provide no evidence of excessive pulsatility as a trigger of BBB disruption.


Subject(s)
Blood-Brain Barrier , Cognition , Magnetic Resonance Imaging , Humans , Blood-Brain Barrier/diagnostic imaging , Aged , Male , Female , Cognition/physiology , Aged, 80 and over , Pulsatile Flow , Cerebral Arteries/diagnostic imaging , Cerebral Arteries/physiology , Prospective Studies , Hippocampus/diagnostic imaging , Hippocampus/physiology , Brain/diagnostic imaging , Brain/physiology , Brain/blood supply , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/diagnostic imaging
3.
Sci Rep ; 14(1): 15317, 2024 07 03.
Article in English | MEDLINE | ID: mdl-38961218

ABSTRACT

The hippocampus is a critical component of the brain and is associated with many neurological disorders. It can be further subdivided into several subfields, and accurate segmentation of these subfields is of great significance for diagnosis and research. However, the structures of hippocampal subfields are irregular and have complex boundaries, and their voxel values are close to surrounding brain tissues, making the segmentation task highly challenging. Currently, many automatic segmentation tools exist for hippocampal subfield segmentation, but they suffer from high time costs and low segmentation accuracy. In this paper, we propose a new dual-branch segmentation network structure (DSnet) based on deep learning for hippocampal subfield segmentation. While traditional convolutional neural network-based methods are effective in capturing hierarchical structures, they struggle to establish long-term dependencies. The DSnet integrates the Transformer architecture and a hybrid attention mechanism, enhancing the network's global perceptual capabilities. Moreover, the dual-branch structure of DSnet leverages the segmentation results of the hippocampal region to facilitate the segmentation of its subfields. We validate the efficacy of our algorithm on the public Kulaga-Yoskovitz dataset. Experimental results indicate that our method is more effective in segmenting hippocampal subfields than conventional single-branch network structures. Compared to the classic 3D U-Net, our proposed DSnet improves the average Dice accuracy of hippocampal subfield segmentation by 0.57%.


Subject(s)
Algorithms , Deep Learning , Hippocampus , Neural Networks, Computer , Hippocampus/diagnostic imaging , Hippocampus/anatomy & histology , Humans , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging/methods
4.
J Mol Neurosci ; 74(3): 61, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38954245

ABSTRACT

Lifestyle influences physical and cognitive development during the period of adolescence greatly. The most important of these lifestyle factors are diet and stress. Therefore, the aim of this study was to investigate the impact of high fat diet (HFD) and chronic mild stress on cognitive function and anxiety-like behaviors in young rats and to study the role of caffeic acid as a potential treatment for anxiety and cognitive dysfunction. Forty rats were assigned into 4 groups: control, HFD, HFD + stress, and caffeic acid-treated group. Rats were sacrificed after neurobehavioral testing. We detected memory impairment and anxiety-like behavior in rats which were more exaggerated in stressed rats. Alongside the behavioral changes, there were biochemical and histological changes. HFD and/or stress decreased hippocampal brain-derived neurotrophic factor (BDNF) levels and induced oxidative and inflammatory changes in the hippocampus. In addition, they suppressed Wnt/ß-catenin pathway which was associated with activation of glycogen synthase kinase 3ß (GSK3ß). HFD and stress increased arginase 1 and inducible nitric oxide synthase (iNOS) levels as well. These disturbances were found to be aggravated in stressed rats than HFD group. However, caffeic acid was able to reverse these deteriorations leading to memory improvement and ameliorating anxiety-like behavior. So, the current study highlights an important neuroprotective role for caffeic acid that may guard against induction of cognitive dysfunction and anxiety disorders in adolescents who are exposed to HFD and/or stress.


Subject(s)
Anxiety , Brain-Derived Neurotrophic Factor , Caffeic Acids , Diet, High-Fat , Glycogen Synthase Kinase 3 beta , Hippocampus , Neuroprotective Agents , Stress, Psychological , Animals , Caffeic Acids/pharmacology , Caffeic Acids/therapeutic use , Rats , Glycogen Synthase Kinase 3 beta/metabolism , Anxiety/drug therapy , Anxiety/etiology , Male , Diet, High-Fat/adverse effects , Hippocampus/metabolism , Hippocampus/drug effects , Stress, Psychological/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Brain-Derived Neurotrophic Factor/metabolism , Rats, Wistar , beta Catenin/metabolism , Wnt Signaling Pathway/drug effects , Cognition/drug effects , Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/drug therapy , Nitric Oxide Synthase Type II/metabolism
5.
Elife ; 122024 Jul 02.
Article in English | MEDLINE | ID: mdl-38953517

ABSTRACT

The hippocampal-dependent memory system and striatal-dependent memory system modulate reinforcement learning depending on feedback timing in adults, but their contributions during development remain unclear. In a 2-year longitudinal study, 6-to-7-year-old children performed a reinforcement learning task in which they received feedback immediately or with a short delay following their response. Children's learning was found to be sensitive to feedback timing modulations in their reaction time and inverse temperature parameter, which quantifies value-guided decision-making. They showed longitudinal improvements towards more optimal value-based learning, and their hippocampal volume showed protracted maturation. Better delayed model-derived learning covaried with larger hippocampal volume longitudinally, in line with the adult literature. In contrast, a larger striatal volume in children was associated with both better immediate and delayed model-derived learning longitudinally. These findings show, for the first time, an early hippocampal contribution to the dynamic development of reinforcement learning in middle childhood, with neurally less differentiated and more cooperative memory systems than in adults.


Subject(s)
Corpus Striatum , Hippocampus , Learning , Reinforcement, Psychology , Humans , Child , Hippocampus/physiology , Longitudinal Studies , Female , Male , Corpus Striatum/physiology , Learning/physiology , Magnetic Resonance Imaging , Decision Making/physiology , Reaction Time/physiology
6.
Nat Commun ; 15(1): 5609, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965228

ABSTRACT

Epilepsy affects 1% of the general population and 30% of patients are resistant to antiepileptic drugs. Although optogenetics is an efficient antiepileptic strategy, the difficulty of illuminating deep brain areas poses translational challenges. Thus, the search of alternative light sources is strongly needed. Here, we develop pH-sensitive inhibitory luminopsin (pHIL), a closed-loop chemo-optogenetic nanomachine composed of a luciferase-based light generator, a fluorescent sensor of intracellular pH (E2GFP), and an optogenetic actuator (halorhodopsin) for silencing neuronal activity. Stimulated by coelenterazine, pHIL experiences bioluminescence resonance energy transfer between luciferase and E2GFP which, under conditions of acidic pH, activates halorhodopsin. In primary neurons, pHIL senses the intracellular pH drop associated with hyperactivity and optogenetically aborts paroxysmal activity elicited by the administration of convulsants. The expression of pHIL in hippocampal pyramidal neurons is effective in decreasing duration and increasing latency of pilocarpine-induced tonic-clonic seizures upon in vivo coelenterazine administration, without affecting higher brain functions. The same treatment is effective in markedly decreasing seizure manifestations in a murine model of genetic epilepsy. The results indicate that pHIL represents a potentially promising closed-loop chemo-optogenetic strategy to treat drug-refractory epilepsy.


Subject(s)
Epilepsy , Neurons , Optogenetics , Animals , Hydrogen-Ion Concentration , Mice , Neurons/metabolism , Neurons/drug effects , Epilepsy/physiopathology , Epilepsy/metabolism , Epilepsy/drug therapy , Humans , Seizures/drug therapy , Seizures/physiopathology , Seizures/metabolism , Halorhodopsins/metabolism , Halorhodopsins/genetics , Hippocampus/metabolism , Hippocampus/drug effects , Male , Luciferases/metabolism , Luciferases/genetics , Pyramidal Cells/metabolism , Pyramidal Cells/drug effects , Imidazoles/pharmacology , Pilocarpine/pharmacology , Disease Models, Animal , Mice, Inbred C57BL , HEK293 Cells , Pyrazines
7.
Transl Psychiatry ; 14(1): 277, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965230

ABSTRACT

The mechanisms contributing to alcohol use disorder (AUD) are complex and the orexigenic peptide ghrelin, which enhances alcohol reward, is implied as a crucial modulator. The major proportion of circulating ghrelin is however the non-octanoylated form of ghrelin, des-acyl ghrelin (DAG), whose role in reward processes is unknown. As recent studies show that DAG decreases food intake, we hypothesize that DAG attenuates alcohol-related responses in animal models. Acute and repeated DAG treatment dose-dependently decreased alcohol drinking in male and female rats. In these alcohol-consuming male rats, repeated DAG treatment causes higher levels of dopamine metabolites in the ventral tegmental area, an area central to reward processing. The role of DAG in reward processing is further supported as DAG prevents alcohol-induced locomotor stimulation, reward in the conditioned place preference paradigm, and dopamine release in the nucleus accumbens in male rodents. On the contrary, DAG does not alter the memory of alcohol reward or affect neurotransmission in the hippocampus, an area central to memory. Further, circulating DAG levels are positively correlated with alcohol drinking in female but not male rats. Studies were conducted in attempts to identify tentative targets of DAG, which currently are unknown. Data from these recombinant cell system revealed that DAG does not bind to either of the monoamine transporters, 5HT2A, CB1, or µ-opioid receptors. Collectively, our data show that DAG attenuates alcohol-related responses in rodents, an effect opposite to that of ghrelin, and contributes towards a deeper insight into behaviors regulated by the ghrelinergic signaling pathway.


Subject(s)
Alcohol Drinking , Dopamine , Ghrelin , Nucleus Accumbens , Reward , Ventral Tegmental Area , Animals , Ghrelin/pharmacology , Ghrelin/metabolism , Male , Rats , Female , Dopamine/metabolism , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/drug effects , Nucleus Accumbens/metabolism , Nucleus Accumbens/drug effects , Ethanol/pharmacology , Ethanol/administration & dosage , Humans , Hippocampus/metabolism , Hippocampus/drug effects , Rats, Sprague-Dawley
8.
Elife ; 122024 Jul 03.
Article in English | MEDLINE | ID: mdl-38958562

ABSTRACT

Hippocampal replay - the time-compressed, sequential reactivation of ensembles of neurons related to past experience - is a key neural mechanism of memory consolidation. Replay typically coincides with a characteristic pattern of local field potential activity, the sharp-wave ripple (SWR). Reduced SWR rates are associated with cognitive impairment in multiple models of neurodegenerative disease, suggesting that a clinically viable intervention to promote SWRs and replay would prove beneficial. We therefore developed a neurofeedback paradigm for rat subjects in which SWR detection triggered rapid positive feedback in the context of a memory-dependent task. This training protocol increased the prevalence of task-relevant replay during the targeted neurofeedback period by changing the temporal dynamics of SWR occurrence. This increase was also associated with neural and behavioral forms of compensation after the targeted period. These findings reveal short-timescale regulation of SWR generation and demonstrate that neurofeedback is an effective strategy for modulating hippocampal replay.


Subject(s)
Hippocampus , Neurofeedback , Animals , Rats , Hippocampus/physiology , Male , Memory Consolidation/physiology , Memory/physiology , Neurons/physiology
9.
Curr Biol ; 34(13): R637-R639, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38981432

ABSTRACT

Memory consolidation is the process of translating memory traces from the hippocampus to the cortex. Hippocampal ripples are key in driving this transfer. A new study now shows that independent cortical ripples can suppress this communication. What could be the underlying mechanisms?


Subject(s)
Hippocampus , Prefrontal Cortex , Hippocampus/physiology , Prefrontal Cortex/physiology , Animals , Memory Consolidation/physiology , Humans , Brain Waves/physiology , Memory/physiology
10.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 36(6): 616-623, 2024 Jun.
Article in Chinese | MEDLINE | ID: mdl-38991961

ABSTRACT

OBJECTIVE: To investigate whether 6-shogaol (6-SH) alleviates oxygen-glucose deprivation/reoxygenation (OGD/R)-induced neuronal autophagy and calcium overload by promoting the expression of microRNA-26a-5p (miR-26a-5p) and inhibiting death-associated protein kinase 1 (DAPK1), and to explore its potential mechanisms. METHODS: Primary cultured logarithmic growth phase mouse hippocampal neurons HT22 cells were taken and cell counting kit-8 (CCK-8) was used to detect cell viability, searching for the optimal concentration of Na2S2O4. HT22 cells were divided into blank control group (NC group), OGD/R group (sugar-free culture medium + 10 mmol/L Na2S2O4 treatment for 1.5 hours followed by normal culture medium for 4 hours), 6-SH intervention group (cultured with 10 µmol/L 6-SH for 4 hours after OGD), negative control inhibitor pretreatment group (transfected with negative control inhibitor for 48 hours followed by OGD, then cultured with 6-SH for 4 hours), and miR-26a-5p inhibitor pretreatment group (transfected with miR-26a-5p inhibitor for 48 hours followed by OGD, then cultured with 6-SH for 4 hours). Cell viability of each group was detected by CCK-8 method; cell ultrastructure was observed under transmission electron microscopy; real-time quantitative polymerase chain reaction (RT-qPCR) was used to detect the gene expressions of DAPK1 and miR-26a-5p; molecular docking were used to verify the interaction between 6-SH and miR-26a-5p; dual-luciferase assay was used to verify the targeting relationship between DAPK1 and miR-26a-5p; flow cytometry was used to determine the levels of intracellular Ca2+; Western blotting was used to detect the protein expressions of phosphorylated-glutamate receptor 2B (p-NMDAR2B) Ser1303, DAPK1, autophagy related protein Beclin1, light chain 3 (LC3), and p-DAPK1 Ser308; immunofluorescence was used to detect the expression of LC3 and Beclin1. RESULTS: The results of the CCK-8 assay showed that the cell viability of the 6-SH intervention group was significantly increased compared to the OGD/R group, while the cell viability of the miR-26a-5p inhibitor pretreatment group was significantly decreased compared to the 6-SH intervention group. Transmission electron microscopy revealed that the number of autophagosomes in the 6-SH intervention group was significantly reduced compared to the OGD/R group, while the number of autophagosomes in the miR-26a-5p inhibitor pretreatment group was significantly increased compared to the 6-SH intervention group. RT-qPCR results showed that compared with the OGD/R group, the expression of miR-26a-5p was significantly upregulated and the expression of DAPK1 mRNA was significantly downregulated in the 6-SH intervention group; compared with the 6-SH intervention group, the expression of miR-26a-5p was significantly downregulated and the expression of DAPK1 mRNA was significantly upregulated in the miR-26a-5p inhibitor pretreatment group. Molecular docking verified the interaction between 6-SH and miR-26a-5p. Dual-luciferase reporter gene assay showed that compared with the negative control group, mmu-miR-26a-5p significantly downregulated the luciferase expression of m-DAPK1-3UTR-WT, indicating a binding interaction between them. Flow cytometry results showed that compared with the OGD/R group, the level of intracellular Ca2+; was significantly decreased in the 6-SH intervention group; compared with the 6-SH intervention group, the level of Ca2+ was significantly increased in the miR-26a-5p inhibitor pretreatment group. Western blotting results showed that compared with the OGD/R group, the protein expressions of p-NMDAR2B Ser1303, DAPK1, Beclin1, and LC3 were significantly decreased in the 6-SH intervention group (p-NMDAR2B Ser1303/ß-actin: 2.34±0.27 vs. 4.78±0.39, DAPK1/ß-actin: 1.40±0.13 vs. 2.37±0.21, Beclin1/ß-actin: 2.61±0.32 vs. 4.32±0.29, LC3/ß-actin: 2.52±0.45 vs. 5.09±0.18, all P < 0.05), while the protein expression of p-DAPK1 Ser308 was significantly increased (p-DAPK1 Ser308/ß-actin: 0.66±0.09 vs. 0.40±0.02, P < 0.05); compared with the 6-SH intervention group, the protein expressions of p-NMDAR2B Ser1303, DAPK1, Beclin1, and LC3 were significantly increased in the miR-26a-5p inhibitor pretreatment group (p-NMDAR2B Ser1303/ß-actin: 4.08±0.14 vs. 2.34±0.27, DAPK1/ß-actin: 1.96±0.15 vs. 1.40±0.13, Beclin1/ß-actin: 3.92±0.31 vs. 2.61±0.32, LC3/ß-actin: 4.33±0.33 vs. 2.52±0.45, all P < 0.05), while the expression of p-DAPK1 Ser308 protein was significantly decreased (p-DAPK1 Ser308/ß-actin: 0.33±0.12 vs. 0.66±0.09, P < 0.05); immunofluorescence staining showed that compared with the OGD/R group, the fluorescence intensity of LC3 and Beclin1 was significantly decreased in the 6-SH intervention group; compared with the 6-SH intervention group, the fluorescence intensity of LC3 and Beclin1 was significantly increased in the miR-26a-5p inhibitor pretreatment group. CONCLUSIONS: 6-SH can alleviate neuronal damage by regulating miR-26a-5p/DAPK1 to reduce autophagy and calcium overload in cells.


Subject(s)
Autophagy , Death-Associated Protein Kinases , MicroRNAs , Reperfusion Injury , MicroRNAs/genetics , Animals , Mice , Death-Associated Protein Kinases/metabolism , Death-Associated Protein Kinases/genetics , Autophagy/drug effects , Neurons/metabolism , Neurons/drug effects , Brain Ischemia/metabolism , Catechols/pharmacology , Cell Survival/drug effects , Hippocampus/metabolism , Glucose
11.
Alzheimers Res Ther ; 16(1): 145, 2024 07 03.
Article in English | MEDLINE | ID: mdl-38961437

ABSTRACT

BACKGROUND: Heat-related illness (HRI) is commonly considered an acute condition, and its potential long-term consequences are not well understood. We conducted a population-based cohort study and an animal experiment to evaluate whether HRI is associated with dementia later in life. METHODS: The Taiwan National Health Insurance Research Database was used in the epidemiological study. We identified newly diagnosed HRI patients between 2001 and 2015, but excluded those with any pre-existing dementia, as the study cohort. Through matching by age, sex, and the index date with the study cohort, we selected individuals without HRI and without any pre-existing dementia as a comparison cohort at a 1:4 ratio. We followed each cohort member until the end of 2018 and compared the risk between the two cohorts using Cox proportional hazards regression models. In the animal experiment, we used a rat model to assess cognitive functions and the histopathological changes in the hippocampus after a heat stroke event. RESULTS: In the epidemiological study, the study cohort consisted of 70,721 HRI patients and the comparison cohort consisted of 282,884 individuals without HRI. After adjusting for potential confounders, the HRI patients had a higher risk of dementia (adjusted hazard ratio [AHR] = 1.24; 95% confidence interval [CI]: 1.19-1.29). Patients with heat stroke had a higher risk of dementia compared with individuals without HRI (AHR = 1.26; 95% CI: 1.18-1.34). In the animal experiment, we found cognitive dysfunction evidenced by animal behavioral tests and observed remarkable neuronal damage, degeneration, apoptosis, and amyloid plaque deposition in the hippocampus after a heat stroke event. CONCLUSIONS: Our epidemiological study indicated that HRI elevated the risk of dementia. This finding was substantiated by the histopathological features observed in the hippocampus, along with the cognitive impairments detected, in the experimental heat stroke rat model.


Subject(s)
Dementia , Animals , Dementia/epidemiology , Dementia/pathology , Male , Female , Humans , Aged , Taiwan/epidemiology , Rats , Cohort Studies , Hippocampus/pathology , Middle Aged , Heat Stress Disorders/epidemiology , Heat Stress Disorders/complications , Aged, 80 and over , Risk Factors , Disease Models, Animal
12.
J Neurosci Res ; 102(7): e25367, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39001670

ABSTRACT

The ventral subiculum regulates emotion, stress responses, and spatial and social cognition. In our previous studies, we have demonstrated anxiety- and depression-like symptoms, deficits in spatial and social cognition in ventral subicular lesioned (VSL) rats, and restoration of affective and cognitive behaviors following photoperiod manipulation (short photoperiod regime, SPR; 6:18 LD cycle). In the present study, we have studied the impact of VSL on sleep-wake behavioral patterns and the effect of SPR on sleep-wakefulness behavior. Adult male Wistar rats subjected to VSL demonstrated decreased wake duration and enhanced total sleep time due to increased non-rapid eye movement sleep (NREMS) and rapid eye movement sleep (REMS). Power spectral analysis indicated increased delta activity during NREMS and decreased sigma band power during all vigilance states. Light is one of the strongest entrainers of the circadian rhythm, and its manipulation may have various physiological and functional consequences. We investigated the effect of 21-day exposure to SPR on sleep-wakefulness (S-W) behavior in VSL rats. We observed that SPR exposure restored S-W behavior in VSL rats, resulting in an increase in wake duration and a significant increase in theta power during wake and REMS. This study highlights the crucial role of the ventral subiculum in maintaining normal sleep-wakefulness patterns and highlights the effectiveness of photoperiod manipulation as a non-pharmacological treatment for reversing sleep disturbances reported in mood and neuropsychiatric disorders like Alzheimer's disease, bipolar disorder, and major depressive disorder, which also involve alterations in circadian rhythm.


Subject(s)
Electroencephalography , Hippocampus , Photoperiod , Rats, Wistar , Sleep , Wakefulness , Animals , Male , Wakefulness/physiology , Rats , Hippocampus/physiopathology , Sleep/physiology , Circadian Rhythm/physiology
13.
Sci Rep ; 14(1): 15855, 2024 Jul 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
14.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(7): 769-775, 2024 Jul 10.
Article in Chinese | MEDLINE | ID: mdl-38946356

ABSTRACT

OBJECTIVE: To identify additional loci associated with depression and the hippocampus (HIP) through genome-wide association study. METHODS: The depression-related genome-wide association study (GWAS) meta summary data was downloaded from the official website of the Psychiatric Genomics Consortium, which had involved 170 756 cases and 329 443 controls. The left and right hippocampal volume GWAS data sets were downloaded from the UK Biobank, which involved 33 224 participants. The conditional false discovery rate (condFDR) was used to identify novel genetic loci for depression and left and right hippocampal volumes, and a conjunctional false discovery rate (conjFDR) was used to evaluate the enrichment of pleiotropic loci between depression and left and right hippocampal volumes. RESULTS: Respectively, 7, 13, and 12 new loci have been associated with depression, left hippocampal volume and right hippocampal volume, with a significant threshold of condFDR < 0.01. A site of rs1267073 locus was found to be shared by the depression and right hippocampal volume with a threshold of conjFDR < 0.01. CONCLUSION: Above findings have provided more insights into the genetic mechanisms underlying the volume of hippocampus and the risk for depression. The results may also provide evidence for future clinical trials for treating depression.


Subject(s)
Depressive Disorder, Major , Genetic Loci , Genome-Wide Association Study , Hippocampus , Humans , Depressive Disorder, Major/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , White People/genetics , Female
15.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(6): 481-487, 2024 Jun.
Article in Chinese | MEDLINE | ID: mdl-38952086

ABSTRACT

Objective To elucidate the role of chaperone-mediated autophagy (CMA) in alleviating emotional dysfunction in mice with sepsis-associated encephalopathy (SAE). Methods The SAE mouse model was established by cecal ligation and perforation (CLP). The severity of sepsis was assessed using the sepsis severity score (MSS). Emotional function in SAE mice was assessed by the open-field test and elevated plus-maze. The expression levels of cognitive heat shock cognate protein 70 (HSC70), lysosomal-associated membrane protein 2A (LAMP2A) and high mobility group box 1 protein B1 (HMGB1) were detected using Western blotting. Co-localization of LAMP2A in the hippocampal neurons was observed by immunofluorescence. The release of inflammatory factors interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α) was measured using ELISA. Following 12 hours post-CLP, mice were orally administered resveratrol at a dose of 30 mg/kg once daily until day 14. Results The mortality rate of CLP mice was 45.83% 24 days post CLP, and all surviving mice exhibited emotional disturbances. 24 hours after CLP, a significant decrease in HSC70 and LAMP2A expression in hippocampal neurons was observed, indicating impaired CMA activity. Meanwhile, HMGB1 and inflammatory cytokines (IL-6 and TNF-α) levels increased. After resveratrol treatment, an increase of HSC70 and LAMP2A expression, and a decrease of HMGB1 expression and inflammatory cytokine release were observed, suggesting enhanced CMA activity and reduced neuroinflammation. Behavioral tests showed that emotional dysfunction was improved in SAE mice after resveratrol treatment. Conclusion CMA activity of hippocampal neurons in SAE mice is significantly reduced, leading to emotional dysfunction. Resveratrol can alleviate neuroinflammation and emotional dysfunction in SAE mice by promoting CMA and inhibiting the expression of HMGB1 and the release of inflammatory factors.


Subject(s)
Chaperone-Mediated Autophagy , HMGB1 Protein , Resveratrol , Sepsis-Associated Encephalopathy , Animals , Mice , Sepsis-Associated Encephalopathy/drug therapy , Sepsis-Associated Encephalopathy/physiopathology , Sepsis-Associated Encephalopathy/metabolism , Male , Resveratrol/pharmacology , HMGB1 Protein/metabolism , Chaperone-Mediated Autophagy/drug effects , Tumor Necrosis Factor-alpha/metabolism , Lysosomal-Associated Membrane Protein 2/metabolism , Lysosomal-Associated Membrane Protein 2/genetics , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Hippocampus/metabolism , Hippocampus/drug effects , Interleukin-6/metabolism , Stilbenes/pharmacology , HSC70 Heat-Shock Proteins/metabolism , Sepsis/complications , Sepsis/drug therapy , Sepsis/metabolism , Sepsis/physiopathology , Mice, Inbred C57BL , Disease Models, Animal
16.
Front Neural Circuits ; 18: 1326609, 2024.
Article in English | MEDLINE | ID: mdl-38947492

ABSTRACT

Gamma oscillations nested in a theta rhythm are observed in the hippocampus, where are assumed to play a role in sequential episodic memory, i.e., memorization and retrieval of events that unfold in time. In this work, we present an original neurocomputational model based on neural masses, which simulates the encoding of sequences of events in the hippocampus and subsequent retrieval by exploiting the theta-gamma code. The model is based on a three-layer structure in which individual Units oscillate with a gamma rhythm and code for individual features of an episode. The first layer (working memory in the prefrontal cortex) maintains a cue in memory until a new signal is presented. The second layer (CA3 cells) implements an auto-associative memory, exploiting excitatory and inhibitory plastic synapses to recover an entire episode from a single feature. Units in this layer are disinhibited by a theta rhythm from an external source (septum or Papez circuit). The third layer (CA1 cells) implements a hetero-associative net with the previous layer, able to recover a sequence of episodes from the first one. During an encoding phase, simulating high-acetylcholine levels, the network is trained with Hebbian (synchronizing) and anti-Hebbian (desynchronizing) rules. During retrieval (low-acetylcholine), the network can correctly recover sequences from an initial cue using gamma oscillations nested inside the theta rhythm. Moreover, in high noise, the network isolated from the environment simulates a mind-wandering condition, randomly replicating previous sequences. Interestingly, in a state simulating sleep, with increased noise and reduced synapses, the network can "dream" by creatively combining sequences, exploiting features shared by different episodes. Finally, an irrational behavior (erroneous superimposition of features in various episodes, like "delusion") occurs after pathological-like reduction in fast inhibitory synapses. The model can represent a straightforward and innovative tool to help mechanistically understand the theta-gamma code in different mental states.


Subject(s)
Gamma Rhythm , Imagination , Models, Neurological , Theta Rhythm , Gamma Rhythm/physiology , Theta Rhythm/physiology , Humans , Imagination/physiology , Memory/physiology , Hippocampus/physiology , Neural Networks, Computer , Animals
17.
Physiol Rep ; 12(13): e16095, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38946616

ABSTRACT

The present study aimed to investigate the effect of catechin-loaded Chitosan-Alginate nanoparticles (NPs) on cognitive function in an aluminum chloride (AlCl3)-induced rat model of Alzheimer's disease (AD). The Catechin-loaded Chitosan-Alginate nanocarriers were synthesized through ionotropic gelation (IG) method. Physio-chemical characterization was conducted with the Zetasizer Nano system, the scanning electron microscope, and the Fourier transform infrared spectroscopy. The experiments were performed over 21 days on six groups of male Wistar rats. The control group, AlCl3 treated group, Catechin group, nanocarrier group, treatment group 1 (AlCl3 + Catechin), and treatment group 2 (AlCl3 + nanocarrier). A behavioral study was done by the Morris water maze (MWM) test. In addition, the level of oxidative indices and acetylcholine esterase (AChE) activity was determined by standard procedures at the end of the study. AlCl3 induced a significant increase in AChE activity, along with a significant decrease in the level of Catalase (CAT) and total antioxidant capacity (TAC) in the hippocampus. Moreover, the significant effect of AlCl3 was observed on the behavioral parameters of the MWM test. Both forms of Catechin markedly improved AChE activity, oxidative biomarkers, spatial memory, and learning. The present study indicated that the administration of Catechin-loaded Chitosan-Alginate NPs is a beneficial therapeutic option against behavioral and chemical alteration of AD in male Wistar rats.


Subject(s)
Alginates , Aluminum Chloride , Alzheimer Disease , Catechin , Chitosan , Nanoparticles , Rats, Wistar , Animals , Catechin/administration & dosage , Catechin/pharmacology , Aluminum Chloride/toxicity , Chitosan/chemistry , Chitosan/administration & dosage , Alginates/chemistry , Alginates/administration & dosage , Male , Alzheimer Disease/drug therapy , Alzheimer Disease/chemically induced , Alzheimer Disease/metabolism , Rats , Administration, Oral , Cognition/drug effects , Acetylcholinesterase/metabolism , Maze Learning/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Disease Models, Animal , Antioxidants/pharmacology , Antioxidants/administration & dosage , Oxidative Stress/drug effects , Drug Carriers/chemistry
18.
Brain Behav ; 14(7): e3586, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38970230

ABSTRACT

BACKGROUND: Patients with myocardial infarction (MI) frequently experience a heightened incidence of depression, thereby increasing the risk of adverse cardiovascular events. Consequently, early detection and intervention in depressive symptoms among patients with MI are imperative. Shexiang Baoxin Pills (SBP), a Chinese patent medicine employed for the treatment of MI, exhibits diverse mechanisms targeting this condition. Nevertheless, its therapeutic efficacy on postmyocardial infarction depressive symptoms remains unclear. The aim of this study is to investigate the effectiveness and mechanism of SBP in managing depression during acute myocardial infarction (AMI). METHODS: A rat model combining MI and depression was established, and the rats were randomly divided into four groups: the model (MOD) group, SBP group, Fluoxetine (FLX) group, and Sham group. After 28 days of drug intervention, cardiac function was assessed using echocardiography while behavior was evaluated through sucrose preference test (SPT), forced swimming test (FST), and open-field test (OFT). Additionally, levels of inflammatory factors in serum and hippocampus were measured along with NLRP3 inflammasome-related protein expression via Western blotting and immunofluorescence. RESULTS: SBP can enhance cardiac function in rats with AMI and depression, while significantly ameliorating depressive-like behavior. Compared to the Sham group, levels of IL-1ß, IL-18, TNF-α, and other inflammatory factors were markedly elevated in the MOD group. However, expressions of these inflammatory factors were reduced to varying degrees following treatment with SBP or FLX. Analysis of NLRP3 inflammasome-related proteins in the hippocampus revealed a significant upregulation of IL-1ß, IL-18, NLRP3, ASC, caspase-1, and GSDMD in the MOD group; conversely, these measures were significantly attenuated after SBP intervention. CONCLUSION: We have observed a significant amelioration in depression-like behavior upon SBP administration during the treatment of AMI, suggesting that this effect may be attributed to the inhibition of NLRP3-mediated pyroptosis. (The main findings are summarized in the graphical abstract in the supplementary file.).


Subject(s)
Antidepressive Agents , Depression , Drugs, Chinese Herbal , Inflammasomes , Myocardial Infarction , NLR Family, Pyrin Domain-Containing 3 Protein , Rats, Sprague-Dawley , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardial Infarction/complications , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/administration & dosage , Rats , Depression/drug therapy , Depression/etiology , Antidepressive Agents/pharmacology , Antidepressive Agents/administration & dosage , Male , Inflammasomes/metabolism , Inflammasomes/drug effects , Disease Models, Animal , Signal Transduction/drug effects , Hippocampus/metabolism , Hippocampus/drug effects , Behavior, Animal/drug effects
19.
Stem Cell Res Ther ; 15(1): 200, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38971770

ABSTRACT

BACKGROUND: Neural stem cell (NSC) proliferation and differentiation in the mammalian brain decreases to minimal levels postnatally. Nevertheless, neurogenic niches persist in the adult cortex and hippocampus in rodents, primates and humans, with adult NSC differentiation sharing key regulatory mechanisms with development. Adult neurogenesis impairments have been linked to Alzheimer's disease (AD) pathology. Addressing these impairments by using neurotrophic factors is a promising new avenue for therapeutic intervention based on neurogenesis. However, this possibility has been hindered by technical difficulties of using in-vivo models to conduct screens, including working with scarce NSCs in the adult brain and differences between human and mouse models or ethical limitations. METHODS: Here, we use a combination of mouse and human stem cell models for comprehensive in-vitro characterization of a novel neurogenic compound, focusing on the brain-derived neurotrophic factor (BDNF) pathway. The ability of ENT-A011, a steroidal dehydroepiandrosterone derivative, to activate the tyrosine receptor kinase B (TrkB) receptor was tested through western blotting in NIH-3T3 cells and its neurogenic and neuroprotective action were assessed through proliferation, cell death and Amyloid-ß (Aß) toxicity assays in mouse primary adult hippocampal NSCs, mouse embryonic cortical NSCs and neural progenitor cells (NPCs) differentiated from three human induced pluripotent stem cell lines from healthy and AD donors. RNA-seq profiling was used to assess if the compound acts through the same gene network as BDNF in human NPCs. RESULTS: ENT-A011 was able to increase proliferation of mouse primary adult hippocampal NSCs and embryonic cortical NSCs, in the absence of EGF/FGF, while reducing Aß-induced cell death, acting selectively through TrkB activation. The compound was able to increase astrocytic gene markers involved in NSC maintenance, protect hippocampal neurons from Αß toxicity and prevent synapse loss after Aß treatment. ENT-A011 successfully induces proliferation and prevents cell death after Aß toxicity in human NPCs, acting through a core gene network shared with BDNF as shown through RNA-seq. CONCLUSIONS: Our work characterizes a novel BDNF mimetic with preferable pharmacological properties and neurogenic and neuroprotective actions in Alzheimer's disease via stem cell-based screening, demonstrating the promise of stem cell systems for short-listing competitive candidates for further testing.


Subject(s)
Alzheimer Disease , Neural Stem Cells , Neurogenesis , Neuroprotective Agents , Receptor, trkB , Animals , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Mice , Neurogenesis/drug effects , Receptor, trkB/metabolism , Receptor, trkB/agonists , Receptor, trkB/genetics , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Neuroprotective Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Amyloid beta-Peptides/metabolism , Hippocampus/drug effects , Hippocampus/metabolism
20.
Stem Cell Res Ther ; 15(1): 197, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38971774

ABSTRACT

BACKGROUND: Traumatic Brain Injury (TBI) represents one of the main causes of brain damage in young people and the elderly population with a very high rate of psycho-physical disability and death. TBI is characterized by extensive cell death, tissue damage and neuro-inflammation with a symptomatology that varies depending on the severity of the trauma from memory loss to a state of irreversible coma and death. Recently, preclinical studies on mouse models have demonstrated that the post-traumatic adult Neural Stem/Progenitor cells response could represent an excellent model to shed light on the neuro-reparative role of adult neurogenesis following damage. The cyclin-dependent kinase inhibitor p21Waf1/Cip1 plays a pivotal role in modulating the quiescence/activation balance of adult Neural Stem Cells (aNSCs) and in restraining the proliferation progression of progenitor cells. Based on these considerations, the aim of this work is to evaluate how the conditional ablation of p21Waf1/Cip1 in the aNSCS can alter the adult hippocampal neurogenesis in physiological and post-traumatic conditions. METHODS: We designed a novel conditional p21Waf1/Cip1 knock-out mouse model, in which the deletion of p21Waf1/Cip1 (referred as p21) is temporally controlled and occurs in Nestin-positive aNSCs, following administration of Tamoxifen. This mouse model (referred as p21 cKO mice) was subjected to Controlled Cortical Impact to analyze how the deletion of p21 could influence the post-traumatic neurogenic response within the hippocampal niche. RESULTS: The data demonstrates that the conditional deletion of p21 in the aNSCs induces a strong increase in activation of aNSCs as well as proliferation and differentiation of neural progenitors in the adult dentate gyrus of the hippocampus, resulting in an enhancement of neurogenesis and the hippocampal-dependent working memory. However, following traumatic brain injury, the increased neurogenic response of aNSCs in p21 cKO mice leads to a fast depletion of the aNSCs pool, followed by declined neurogenesis and impaired hippocampal functionality. CONCLUSIONS: These data demonstrate for the first time a fundamental role of p21 in modulating the post-traumatic hippocampal neurogenic response, by the regulation of the proliferative and differentiative steps of aNSCs/progenitor populations after brain damage.


Subject(s)
Brain Injuries, Traumatic , Cyclin-Dependent Kinase Inhibitor p21 , Hippocampus , Mice, Knockout , Neural Stem Cells , Neurogenesis , Animals , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Neural Stem Cells/metabolism , Mice , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/genetics , Hippocampus/metabolism , Hippocampus/pathology , Disease Models, Animal , Male , Cell Proliferation , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...