Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.463
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
Prog Neurobiol ; 237: 102612, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38642602

ABSTRACT

Recurrent seizures lead to accumulation of the activity-dependent transcription factor ∆FosB in hippocampal dentate granule cells in both mouse models of epilepsy and mouse models of Alzheimer's disease (AD), which is also associated with increased incidence of seizures. In patients with AD and related mouse models, the degree of ∆FosB accumulation corresponds with increasing severity of cognitive deficits. We previously found that ∆FosB impairs spatial memory in mice by epigenetically regulating expression of target genes such as calbindin that are involved in synaptic plasticity. However, the suppression of calbindin in conditions of neuronal hyperexcitability has been demonstrated to provide neuroprotection to dentate granule cells, indicating that ∆FosB may act over long timescales to coordinate neuroprotective pathways. To test this hypothesis, we used viral-mediated expression of ∆JunD to interfere with ∆FosB signaling over the course of several months in transgenic mice expressing mutant human amyloid precursor protein (APP), which exhibit spontaneous seizures and develop AD-related neuropathology and cognitive deficits. Our results demonstrate that persistent ∆FosB activity acts through discrete modes of hippocampal target gene regulation to modulate neuronal excitability, limit recurrent seizure activity, and provide neuroprotection to hippocampal dentate granule cells in APP mice.


Subject(s)
Amyloid beta-Protein Precursor , Dentate Gyrus , Mice, Transgenic , Proto-Oncogene Proteins c-fos , Seizures , Animals , Dentate Gyrus/metabolism , Mice , Seizures/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/genetics , Neuroprotection/physiology , Disease Models, Animal , Alzheimer Disease/metabolism , Male , Mice, Inbred C57BL , Humans
12.
Biomolecules ; 14(3)2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38540798

ABSTRACT

Although granule cell dispersion (GCD) in the hippocampus is known to be an important feature associated with epileptic seizures in temporal lobe epilepsy (TLE), the endogenous molecules that regulate GCD are largely unknown. In the present study, we have examined whether there is any change in AEG-1 expression in the hippocampus of a kainic acid (KA)-induced mouse model of TLE. In addition, we have investigated whether the modulation of astrocyte elevated gene-1 (AEG-1) expression in the dentate gyrus (DG) by intracranial injection of adeno-associated virus 1 (AAV1) influences pathological phenotypes such as GCD formation and seizure susceptibility in a KA-treated mouse. We have identified that the protein expression of AEG-1 is upregulated in the DG of a KA-induced mouse model of TLE. We further demonstrated that AEG-1 upregulation by AAV1 delivery in the DG-induced anticonvulsant activities such as the delay of seizure onset and inhibition of spontaneous recurrent seizures (SRS) through GCD suppression in the mouse model of TLE, while the inhibition of AEG-1 expression increased susceptibility to seizures. The present observations suggest that AEG-1 is a potent regulator of GCD formation and seizure development associated with TLE, and the significant induction of AEG-1 in the DG may have therapeutic potential against epilepsy.


Subject(s)
Epilepsy, Temporal Lobe , Epilepsy , Animals , Mice , Astrocytes/metabolism , Dentate Gyrus/metabolism , Epilepsy/metabolism , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/genetics , Epilepsy, Temporal Lobe/drug therapy , Hippocampus/metabolism , Kainic Acid/adverse effects , Kainic Acid/metabolism , Seizures/chemically induced , Seizures/genetics , Seizures/metabolism
13.
Sci Rep ; 14(1): 6915, 2024 03 22.
Article in English | MEDLINE | ID: mdl-38519576

ABSTRACT

Neuronal aging may be, in part, related to a change in DNA methylation. Thus, methyl donors, like folate and methionine, may play a role in cognitive changes associated to neuronal aging. To test the role of these metabolites, we performed stereotaxic microinjection of these molecules into the dentate gyrus (DG) of aged mice (an average age of 21 month). Folate, but not S-Adenosyl-Methionine (SAM), enhances cognition in aged mice. In the presence of folate, we observed partial rejuvenation of DG cells, characterized by the expression of juvenile genes or reorganization of extracellular matrix. Here, we have also tried to identify the mechanism independent of DNA methylation, that involve folate effects on cognition. Our analyses indicated that folate binds to folate receptor α (FRα) and, upon folate binding, FRα is transported to cell nucleus, where it is acting as transcription factor for expressing genes like SOX2 or GluN2B. In this work, we report that a FRα binding peptide also replicates the folate effect on cognition, in aged mice. Our data suggest that such effect is not sex-dependent. Thus, we propose the use of this peptide to improve cognition since it lacks of folate-mediated side effects. The use of synthetic FRα binding peptides emerge as a future strategy for the study of brain rejuvenation.


Subject(s)
Folate Receptor 1 , Rejuvenation , Animals , Mice , Cognition , Dentate Gyrus/metabolism , Folate Receptor 1/metabolism , Folic Acid/metabolism , Methionine , Peptides/metabolism , S-Adenosylmethionine
14.
Cell Biochem Funct ; 42(2): e3958, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38396357

ABSTRACT

Iron accumulation in the brain causes oxidative stress, blood-brain barrier (BBB) breakdown, and neurodegeneration. We examined the preventive effects of acetylated oligopeptides (AOP) from whey protein on iron-induced hippocampal damage compared to N-acetyl cysteine (NAC). This 5-week study used 40 male albino rats. At the start, all rats received 150 mg/kg/day of oral NAC for a week. The 40 animals were then randomly divided into four groups: Group I (control) received a normal diet; Group II (iron overload) received 60 mg/kg/day intraperitoneal iron dextran 5 days a week for 4 weeks; Group III (NAC group) received 150 mg/kg/day NAC and iron dextran; and Group IV (AOP group) received 150 mg/kg/day AOP and iron dextran. Enzyme-linked immunosorbent assay, spectrophotometry, and qRT-PCR were used to measure MMP-9, tissue inhibitor metalloproteinase-1 (TIMP-1), MDA, reduced glutathione (GSH) levels, and nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) gene expression. Histopathological and immunohistochemical detection of nestin, claudin, caspase, and GFAP was also done. MMP-9, TIMP-1, MDA, caspase, and GFAP rose in the iron overload group, while GSH, Nrf2, HO-1, nestin, and claudin decreased. The NAC and AOP administrations improved iron overload-induced biochemical and histological alterations. We found that AOP and NAC can protect the brain hippocampus from iron overload, improve BBB disruption, and provide neuroprotection with mostly no significant difference from healthy controls.


Subject(s)
Acetylcysteine , Iron Overload , Oligopeptides , Animals , Male , Rats , Acetylcysteine/pharmacology , Acetylcysteine/metabolism , Caspases/metabolism , Claudins/genetics , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Dextrans/metabolism , Dextrans/pharmacology , Down-Regulation , Glutathione/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Iron/metabolism , Iron/pharmacology , Iron Overload/complications , Iron Overload/drug therapy , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/pharmacology , Nestin/genetics , Nestin/metabolism , Nestin/pharmacology , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-1/pharmacology , Up-Regulation , Oligopeptides/pharmacology , Heme Oxygenase-1/drug effects , Glial Fibrillary Acidic Protein/drug effects , Glial Fibrillary Acidic Protein/metabolism
15.
Cell Rep ; 43(3): 113806, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38377001

ABSTRACT

Experience-driven alterations in neuronal activity are followed by structural-functional modifications allowing cells to adapt to these activity changes. Structural plasticity has been observed for cortical principal cells. However, how GABAergic interneurons respond to experience-dependent network activity changes is not well understood. We show that parvalbumin-expressing interneurons (PVIs) of the dentate gyrus (DG) possess dendritic spines, which undergo behaviorally induced structural dynamics. Glutamatergic inputs at PVI spines evoke signals with high spatial compartmentalization defined by neck length. Mice experiencing novel contexts form more PVI spines with elongated necks and exhibit enhanced network and PVI activity and cFOS expression. Enhanced green fluorescent protein reconstitution across synaptic partner-mediated synapse labeling shows that experience-driven PVI spine growth boosts targeting of PVI spines over shafts by glutamatergic synapses. Our findings propose a role for PVI spine dynamics in regulating PVI excitation by their inputs, which may allow PVIs to dynamically adjust their functional integration in the DG microcircuitry in relation to network computational demands.


Subject(s)
Interneurons , Parvalbumins , Mice , Animals , Parvalbumins/metabolism , Interneurons/metabolism , Neurons/metabolism , Synapses/metabolism , Dentate Gyrus/metabolism , Neuronal Plasticity
16.
Pflugers Arch ; 476(5): 821-831, 2024 May.
Article in English | MEDLINE | ID: mdl-38416255

ABSTRACT

Obesity has been linked with the impairment of spatial memory and synaptic plasticity but the molecular mechanisms remained unidentified. Since glutamatergic transmission and NMDA receptor neural pathways in hippocampal dentate gyrus (DG) are essential in the learning and memory, we aimed to investigate glutamate (Glu) and NMDA receptor signaling of DG in spatial learning and memory in diet-induced obesity (DIO) rats. Spatial learning and memory were assessed via Morris water maze (MWM) test on control (Ctr) and DIO rats. Extracellular concentration of Glu in the DG was determined using in vivo microdialysis and HPLC. The protein expressions of NMDA receptor subunit 2B (NR2B), brain-derived neurotrophic factor (BDNF), the activation of calcium/calmodulin-dependent kinase II (CaMKII) and cAMP-response-element-binding protein (CREB) in the DG were observed by western blot. Spatial learning and memory were impaired in DIO rats compared to those of Ctr. NR2B expression was increased, while BDNF expression and CaMKII and CREB activation were decreased in DG of DIO rats. Extracellular concentration of Glu was increased in Ctr on the 3rd and 4th days of the MWM test, but significant further increment was observed in DIO rats. Microinjection of an NMDA antagonist (MK-801) into the DG reversed spatial learning and memory impairment. Such effects were accompanied by greater BDNF expression and CaMKII/CREB activation in the DG of DIO rats. In conclusion, the enhancement of Glu-NMDA receptor transmission in the hippocampal DG contributes to the impairment of spatial learning and memory in DIO rats, maybe via the modulation of CaMKII-CREB-BDNF signaling pathway.


Subject(s)
Dentate Gyrus , Glutamic Acid , Obesity , Receptors, N-Methyl-D-Aspartate , Spatial Learning , Animals , Male , Rats , Brain-Derived Neurotrophic Factor/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Dentate Gyrus/metabolism , Glutamic Acid/metabolism , Maze Learning , Memory Disorders/metabolism , Memory Disorders/etiology , Obesity/metabolism , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/physiology , Spatial Memory , Synaptic Transmission
17.
Behav Brain Res ; 463: 114914, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38368953

ABSTRACT

Previous studies have shown that various receptors, including dopamine receptors, are expressed in the hippocampal dentate gyrus (DG). Besides, indicatively, dopamine receptors play an essential role in the modulation of pain perception. On the other hand, stressful experiences can produce analgesia, termed stress-induced analgesia (SIA). The current study examined the probable role of dopamine receptors within the DG in antinociception induced by restraint stress (RS). Ninety-seven male albino Wistar rats were unilaterally implanted with a cannula in the DG. Animals received intra-DG microinjections of SCH23390 or Sulpiride (0.25, 1, and 4 µg/rat) as D1-and D2-like dopamine receptor antagonists, respectively, five minutes before RS. Ten minutes after the end of the induction of RS for three hours, 50 µl 2.5% formalin was injected subcutaneously into the plantar surface of the hind paw to induce persistent inflammatory pain. Pain scores were evaluated at 5-minute intervals for 60 minutes. These findings showed that; exposure to RS for three hours produced SIA in both phases of the formalin test, while this RS-induced analgesia was attenuated in the early and late phases of the formalin test by intra-DG microinjection of SCH23390 and Sulpiride. The results of the present study suggested that both D1- and D2-like dopamine receptors in the DG have a considerable role in the induced analgesia by RS.


Subject(s)
Receptors, Dopamine , Sulpiride , Rats , Male , Animals , Sulpiride/pharmacology , Pain Measurement , Receptors, Dopamine/physiology , Analgesics/adverse effects , Pain/chemically induced , Rats, Wistar , Dentate Gyrus/metabolism , Hippocampus/metabolism , Receptors, Dopamine D1/metabolism , Benzazepines/pharmacology
18.
J Chem Neuroanat ; 136: 102391, 2024 03.
Article in English | MEDLINE | ID: mdl-38219812

ABSTRACT

BACKGROUND: Maternal diabetes during pregnancy can affect the neurological development of offspring. Glial cell-derived neurotrophic factor (GDNF), neurturin (NRTN), and neural cell adhesion molecules (NCAM) are three important proteins for brain development. Therefore, this study aimed to investigate the impacts of the mentioned neurotrophic factors in the hippocampal dentate gyrus (DG) of rat offspring born to diabetic mothers. METHODS: Wistar female rats were randomly allocated into diabetic (STZ-D) [(45 mg/kg BW, STZ (Streptozotocin), i.p)], diabetic + NPH insulin (STZ-INS) [(4-6 unit/kg/day SC)], and control groups. The animals in all groups were mated by non-diabetic male rats. Two weeks after birth, male pups from each group were sacrificed and then protein contents of GDNF, NRTN, and NCAM were evaluated using immunohistochemistry. RESULTS: The study found that the expression of GDNF and NRTN in the hippocampus of diabetic rat offspring was significantly higher compared to the diabetic+ insulin and control groups, respectively (P < 0.01, P < 0.001). Additionally, the expression of NCAM was significantly higher in the diabetic group the diabetic+ insulin and control groups (P < 0.01, P < 0.001). CONCLUSIONS: The results of the study revealed that diabetes during pregnancy significantly impacts the distribution pattern of GDNF, NRTN, and NCAM in the hippocampus of rat neonates.


Subject(s)
Diabetes, Gestational , Insulins , Humans , Pregnancy , Rats , Animals , Male , Female , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Neurturin/metabolism , Neurturin/pharmacology , Rats, Wistar , Neural Cell Adhesion Molecules/metabolism , Dentate Gyrus/metabolism
19.
Neuroendocrinology ; 114(4): 400-410, 2024.
Article in English | MEDLINE | ID: mdl-38171345

ABSTRACT

INTRODUCTION: Thyroid hormones, which produce critical changes in our bodies even when their physiological levels alter slightly, are crucial hormones that influence gene transcription. Neuronal plasticity, on the other hand, requires both the activation of local proteins as well as protein translation and transcription in response to external signals. So far, no study has examined metaplastic long-term potentiation (LTP) and related gene expression levels in a hyperthyroid experimental model. METHODS: The Wistar male rats were administered 0.2 mg/kg/day of l-thyroxine for 21 days to induce hyperthyroidism. Perforant path was primed with 1-Hz low-frequency stimuli (LFS) for 900 s to investigate metaplasticity responses. The LFS was followed by high-frequency stimuli (HFS, 100 Hz) after 5 min. Excitatory postsynaptic potential (EPSP) slope and population spike (PS) amplitude were recorded from the granule cell layer of the dentate gyrus. The mRNA levels of genes related to neurodegeneration (Gsk-3ß, Cdk5, Akt1, Mapt, p35, Capn1, Bace1, and Psen2) were measured using the RT-PCR method for the stimulated hippocampus. RESULTS: Similar to euthyroid rats, hyperthyroid animals had a lower EPSP slope and PS after LFS. Depression of EPSP prevented subsequently induced EPSP-LTP, although HFS was able to elicit PS-LTP despite depression of PS amplitude in both groups. Despite similarities in metaplastic LTP responses, these electrophysiological findings were accompanied by increased Akt, Bace1, Cdk5, and p35-mRNA expressions and decreased Gsk-3ß mRNA expression in hyperthyroid rats' hippocampus. CONCLUSION: These data support the view that in thyroid hormone excess, the mechanism that keeps synaptic efficacy within a dynamic range occurs concurrently with increased mRNA expression of neurodegeneration-related genes. Our study encourages further examination of the increased risk of neurodegenerative disease in hyperthyroidism.


Subject(s)
Hyperthyroidism , Neurodegenerative Diseases , Rats , Male , Animals , Rats, Wistar , Amyloid Precursor Protein Secretases/adverse effects , Amyloid Precursor Protein Secretases/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Up-Regulation , Neurodegenerative Diseases/metabolism , Aspartic Acid Endopeptidases/adverse effects , Aspartic Acid Endopeptidases/metabolism , Hippocampus/metabolism , Neuronal Plasticity/physiology , Hyperthyroidism/chemically induced , Hyperthyroidism/metabolism , RNA, Messenger/metabolism , Gene Expression , Dentate Gyrus/metabolism
20.
Development ; 151(4)2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38265146

ABSTRACT

Lysosomes are intracellular organelles responsible for degrading diverse macromolecules delivered from several pathways, including the endo-lysosomal and autophagic pathways. Recent reports have suggested that lysosomes are essential for regulating neural stem cells in developing, adult and aged brains. However, the activity of these lysosomes has yet to be monitored in these brain tissues. Here, we report the development of a new probe to measure lysosomal protein degradation in brain tissue by immunostaining. Our results indicate that lysosomal protein degradation fluctuates in neural stem cells of the hippocampal dentate gyrus, depending on age and brain disorders. Neural stem cells increase their lysosomal activity during hippocampal development in the dentate gyrus, but aging and aging-related disease reduce lysosomal activity. In addition, physical exercise increases lysosomal activity in neural stem cells and astrocytes in the dentate gyrus. We therefore propose that three different stages of lysosomal activity exist: the state of increase during development, the stable state during adulthood and the state of reduction due to damage caused by either age or disease.


Subject(s)
Dentate Gyrus , Neural Stem Cells , Animals , Mice , Dentate Gyrus/metabolism , Proteolysis , Neural Stem Cells/metabolism , Astrocytes/metabolism , Lysosomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...