Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(10)2024 May 08.
Article in English | MEDLINE | ID: mdl-38791161

ABSTRACT

Adult neurogenesis in the dentate gyrus (DG) is impaired during Alzheimer's disease (AD) progression. Curcumin has been reported to reduce cell apoptosis and stimulate neurogenesis. This study aimed to investigate the influence of curcumin on adult neurogenesis in AD mice and its potential mechanism. Two-month-old male C57BL/6J mice were injected with soluble ß-amyloid (Aß1-42) using lateral ventricle stereolocalization to establish AD models. An immunofluorescence assay, including bromodeoxyuridine (BrdU), doublecortin (DCX), and neuron-specific nuclear antigen (NeuN), was used to detect hippocampal neurogenesis. Western blot and an enzyme-linked immunosorbent assay (ELISA) were used to test the expression of related proteins and the secretion of brain-derived neurotrophic factor (BDNF). A Morris water maze was used to detect the cognitive function of the mice. Our results showed that curcumin administration (100 mg/kg) rescued the impaired neurogenesis of Aß1-42 mice, shown as enhanced BrdU+/DCX+ and BrdU+/NeuN+ cells in DG. In addition, curcumin regulated the phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt) -mediated glycogen synthase kinase-3ß (GSK3ß) /Wingless/Integrated (Wnt)/ß-catenin pathway and cyclic adenosine monophosphate response element-binding protein (CREB)/BDNF in Aß1-42 mice. Inhibiting Wnt/ß-catenin and depriving BDNF could reverse both the upregulated neurogenesis and cognitive function of curcumin-treated Aß1-42 mice. In conclusion, our study indicates that curcumin, through targeting PI3K/Akt, regulates GSK3ß/Wnt/ß-catenin and CREB/BDNF pathways, improving the adult neurogenesis of AD mice.


Subject(s)
Alzheimer Disease , Curcumin , Neurogenesis , Wnt Signaling Pathway , Animals , Male , Mice , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , beta Catenin/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Curcumin/pharmacology , Disease Models, Animal , Doublecortin Protein/metabolism , Hippocampus/metabolism , Hippocampus/drug effects , Mice, Inbred C57BL , Neurogenesis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Up-Regulation/drug effects , Wnt Signaling Pathway/drug effects
2.
Cells ; 11(1)2022 01 04.
Article in English | MEDLINE | ID: mdl-35011720

ABSTRACT

Williams syndrome (WS) is a multisystem neurodevelopmental disorder caused by a de novo hemizygous deletion of ~26 genes from chromosome 7q11.23, among them the general transcription factor II-I (GTF2I). By studying a novel murine model for the hypersociability phenotype associated with WS, we previously revealed surprising aberrations in myelination and cell differentiation properties in the cortices of mutant mice compared to controls. These mutant mice had selective deletion of Gtf2i in the excitatory neurons of the forebrain. Here, we applied diffusion magnetic resonance imaging and fiber tracking, which showed a reduction in the number of streamlines in limbic outputs such as the fimbria/fornix fibers and the stria terminalis, as well as the corpus callosum of these mutant mice compared to controls. Furthermore, we utilized next-generation sequencing (NGS) analysis of cortical small RNAs' expression (RNA-Seq) levels to identify altered expression of microRNAs (miRNAs), including two from the miR-34 cluster, known to be involved in prominent processes in the developing nervous system. Luciferase reporter assay confirmed the direct binding of miR-34c-5p to the 3'UTR of PTPRU-a gene involved in neural development that was elevated in the cortices of mutant mice relative to controls. Moreover, we found an age-dependent variation in the expression levels of doublecortin (Dcx)-a verified miR-34 target. Thus, we demonstrate the substantial effect a single gene deletion can exert on miRNA regulation and brain structure, and advance our understanding and, hopefully, treatment of WS.


Subject(s)
Brain/growth & development , Doublecortin Protein/metabolism , MicroRNAs/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , White Matter/physiopathology , Williams Syndrome/genetics , Animals , Disease Models, Animal , Female , Humans , Mice , Williams Syndrome/pathology
3.
Int J Mol Sci ; 22(24)2021 Dec 08.
Article in English | MEDLINE | ID: mdl-34948016

ABSTRACT

Glioblastoma (GBM) remains the leading cause of cancer-related deaths with the lowest five-year survival rates among all of the human cancers. Multiple factors contribute to its poor outcome, including intratumor heterogeneity, along with migratory and invasive capacities of tumour cells. Over the last several years Doublecortin (DCX) has been one of the debatable factors influencing GBM cells' migration. To resolve DCX's ambiguous role in GBM cells' migration, we set to analyse the expression patterns of DCX along with Nestin (NES) and Oligodendrocyte lineage transcription factor 2 (OLIG2) in 17 cases of GBM, using immunohistochemistry, followed by an analysis of single-cell RNA-seq data. Our results showed that only a small subset of DCX positive (DCX+) cells was present in the tumour. Moreover, no particular pattern emerged when analysing DCX+ cells relative position to the tumour margin. By looking into single-cell RNA-seq data, the majority of DCX+ cells were classified as non-cancerous, with a small subset of cells that could be regarded as glioma stem cells. In conclusion, our findings support the notion that glioma cells express DCX; however, there is no clear evidence to prove that DCX participates in GBM cell migration.


Subject(s)
Brain Neoplasms/metabolism , Doublecortin Protein/metabolism , Gene Expression Profiling/methods , Glioblastoma/metabolism , Nestin/metabolism , Oligodendrocyte Transcription Factor 2/metabolism , Brain Neoplasms/genetics , Cell Movement , Doublecortin Protein/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Heuristics , Humans , Image Processing, Computer-Assisted , Microscopy, Confocal , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Nestin/genetics , Oligodendrocyte Transcription Factor 2/genetics , Sequence Analysis, RNA , Single-Cell Analysis , Survival Analysis
4.
Sci Rep ; 11(1): 21499, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34728755

ABSTRACT

Adolescence represents a crucial period for maturation of brain structures involved in cognition. Early in life unhealthy dietary patterns are associated with inferior cognitive outcomes at later ages; conversely, healthy diet is associated with better cognitive results. In this study we analyzed the effects of a short period of hypercaloric diet on newborn hippocampal doublecortin+ (DCX) immature neurons in adolescent mice. Male mice received high fat diet (HFD) or control low fat diet (LFD) from the 5th week of age for 1 or 2 weeks, or 1 week HFD followed by 1 week LFD. After diet supply, mice were either perfused for immunohistochemical (IHC) analysis or their hippocampi were dissected for biochemical assays. Detailed morphometric analysis was performed in DCX+ cells that displayed features of immature neurons. We report that 1 week-HFD was sufficient to dramatically reduce dendritic tree complexity of DCX+ cells. This effect occurred specifically in dorsal and not ventral hippocampus and correlated with reduced BDNF expression levels in dorsal hippocampus. Both structural and biochemical changes were reversed by a return to LFD. Altogether these studies increase our current knowledge on potential consequences of hypercaloric diet on brain and in particular on dorsal hippocampal neuroplasticity.


Subject(s)
Diet, High-Fat/adverse effects , Doublecortin Protein/metabolism , Hippocampus/pathology , Neural Stem Cells/pathology , Neurogenesis , Neuronal Plasticity , Neurons/pathology , Animals , Body Weight , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Neurons/metabolism
5.
Int J Mol Sci ; 22(22)2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34830044

ABSTRACT

BMPs regulate synovial quiescence and adult neurogenesis in the hippocampus in non-stress conditions. However, changes in BMP expression that are induced by inflammation during rheumatoid arthritis (RA) have not yet been reported. Here, we show that signalling with synovial BMPs (BMP-4 and -7) mediates the effect of systemic inflammation on adult neurogenesis in the hippocampus during pristane-induced arthritis (PIA) in Dark Agouti (DA) rats, an animal model of RA. Moreover, we show gender differences in BMP expressions and their antagonists (Noggin and Gremlin) during PIA and their correlations with the clinical course and IL-17A and TNF-α levels in serum. Our results indicate gender differences in the clinical course, where male rats showed earlier onset and earlier recovery but a worse clinical course in the first two phases of the disease (onset and peak), which correlates with the initial increase of serum IL-17A level. The clinical course of the female rats worsened in remission. Their prolonged symptoms could be a reflection of an increased TNF-α level in serum during remission. Synovial inflammation was greater in females in PIA-remission with greater synovial BMP and antagonist expressions. More significant correlations between serum cytokines (IL-17A and TNF-α), and synovial BMPs and their antagonists were found in females than in males. On the other hand, males showed an increase in hippocampal BMP-4 expression during the acute phase, but both genders showed a decrease in antagonist expressions during PIA in general. Both genders showed a decrease in the number of Ki-67+ and SOX-2+ and DCX+ cells and in the ratio of DCX+ to Ki67+ cells in the dentate gyrus during PIA. However, in PIA remission, females showed a faster increase in the number of Ki67+, SOX-2+, and DCX+ cells and a faster increase in the DCX/Ki67 ratio than males. Both genders showed an increase of hippocampal BMP-7 expression during remission, although males constantly showed greater BMP-7 expression at all time points. Our data show that gender differences exist in the BMP expressions in the periphery-hippocampus axis and in the IL-17A and TNF-α levels in serum, which could imply differences in the mechanisms for the onset and progression of the disease, the clinical course severity, and adult neurogenesis with subsequent neurological complications between genders.


Subject(s)
Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Bone Morphogenetic Proteins/metabolism , Hippocampus/metabolism , Joints/metabolism , Neurogenesis , Aging , Animals , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 7/metabolism , Carrier Proteins/metabolism , Cytokines/metabolism , Doublecortin Protein/metabolism , Female , Inflammation/chemically induced , Inflammation/metabolism , Interleukin-17/blood , Ki-67 Antigen/metabolism , Male , Rats , SOXB1 Transcription Factors/metabolism , Sex Factors , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Terpenes/toxicity , Tumor Necrosis Factor-alpha/blood
6.
Neuropharmacology ; 201: 108834, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34637786

ABSTRACT

Inflammation is a crucial component that contributes to the pathogenesis of major depressive disorder. It has been revealed that the nonselective cation channel transient receptor potential vanilloid 4 (TRPV4) profoundly affects a variety of physiological processes, including inflammation. However, its roles and mechanisms in LPS-induced depression are still unclear. Here, for the first time, we found that there was a significant increase in TRPV4 in the hippocampus in a depression mouse model induced by LPS. TRPV4 inhibitor HC067047 or knockdown the hippocampal TRPV4 with TRPV4 shRNA could effectively rescue the aberrant behaviors. Furthermore, TRPV4 inhibitor HC067047 reduced the activation of astrocyte and microglia, decreased expression of CaMKII-NLRP3 inflammasome and increased the expression of neurogenesis marker DCX in the hippocampus. In addition, enhanced neuroinflammation in the serum was also reversed by TRPV4 inhibitor HC067047. Thus, we consider that TRPV4 has an important role in contributing to the depression-like behavior following LPS-induced systemic inflammation.


Subject(s)
Antidepressive Agents , Depression , Lipopolysaccharides , Pyrroles , TRPV Cation Channels , Animals , Male , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Astrocytes/metabolism , Depression/chemically induced , Depression/drug therapy , Disease Models, Animal , Doublecortin Protein/metabolism , Hippocampus/metabolism , Inflammasomes/metabolism , Lipopolysaccharides/adverse effects , Mice, Inbred C57BL , Microglia/metabolism , Neurogenesis/genetics , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism , TRPV Cation Channels/physiology , Pyrroles/chemistry , Pyrroles/pharmacology , Pyrroles/therapeutic use
7.
Int J Mol Sci ; 22(20)2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34681636

ABSTRACT

Depression is a neuropsychiatric disorder with a high impact on the worldwide population. To overcome depression, antidepressant drugs are the first line of treatment. However, pre-clinical studies have pointed out that antidepressants are not entirely efficacious and that the quality of the living environment after stress cessation may play a relevant role in increasing their efficacy. As it is unknown whether a short daily exposure to environmental enrichment during chronic stress and antidepressant treatment will be more effective than just the pharmacological treatment, this study analyzed the effects of fluoxetine, environmental enrichment, and their combination on depressive-associated behavior. Additionally, we investigated hippocampal neurogenesis in mice exposed to chronic mild stress. Our results indicate that fluoxetine reversed anhedonia. Besides, fluoxetine reversed the decrement of some events of the hippocampal neurogenic process caused by chronic mild stress. Conversely, short daily exposure to environmental enrichment changed the deterioration of the coat and anhedonia. Although, this environmental intervention did not produce significant changes in the neurogenic process affected by chronic mild stress, fluoxetine plus environmental enrichment showed similar effects to those caused by environmental enrichment to reverse depressive-like behaviors. Like fluoxetine, the combination reversed the declining number of Ki67, doublecortin, calretinin cells and mature newborn neurons. Finally, this study suggests that short daily exposure to environmental enrichment improves the effects of fluoxetine to reverse the deterioration of the coat and anhedonia in chronically stressed mice. In addition, the combination of fluoxetine with environmental enrichment produces more significant effects than those caused by fluoxetine alone on some events of the neurogenic process. Thus, environmental enrichment improves the benefits of pharmacological treatment by mechanisms that need to be clarified.


Subject(s)
Anhedonia/drug effects , Fluoxetine/pharmacology , Hippocampus/drug effects , Neurogenesis/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Stress, Psychological/physiopathology , Anhedonia/physiology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Calbindin 2/metabolism , Cell Proliferation , Doublecortin Protein/metabolism , Environment , Female , Hippocampus/metabolism , Hippocampus/pathology , Ki-67 Antigen/metabolism , Mice , Mice, Inbred BALB C , Stress, Physiological
8.
Biomed Pharmacother ; 144: 112273, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34700232

ABSTRACT

Neurogenesis in the adult brain is well recognized and plays a critical role in the maintenance of brain function and homeostasis. However, whether neurogenesis also occurs in the adult peripheral nervous system remains unknown. Here, using sensory ganglia (dorsal root ganglia, DRGs) as a model, we show that neurogenesis also occurs in the peripheral nervous system, but in a manner different from that in the central nervous system. Satellite glial cells (SGCs) express the neuronal precursor markers Nestin, POU domain, class 4, transcription factor 1, and p75 pan-neurotrophin receptor. Following sciatic nerve injury, the suppression of endogenous proBDNF by proBDNF antibodies resulted in the transformation of proliferating SGCs into doublecortin-positive cells in the DRGs. Using purified SGCs migrating out from the DRGs, the inhibition of endogenous proBDNF promoted the conversion of SGCs into neuronal phenotypes in vitro. Our findings suggest that SGCs are neuronal precursors, and that proBDNF maintains the SGC phenotype. Furthermore, the suppression of proBDNF signaling is necessary for neuronal phenotype acquisition by SGCs. Thus, we propose that peripheral neurogenesis may occur via the direct conversion of SGCs into neurons, and that this process is negatively regulated by proBDNF.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Ganglia, Spinal/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Neuroglia/metabolism , Peripheral Nerve Injuries/metabolism , Protein Precursors/metabolism , Action Potentials , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor/genetics , Cell Transdifferentiation , Cells, Cultured , Disease Models, Animal , Doublecortin Protein/metabolism , Female , Ganglia, Spinal/pathology , Ganglia, Spinal/physiopathology , Male , Neural Stem Cells/pathology , Neuroglia/pathology , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/physiopathology , Phenotype , Protein Precursors/genetics , Rats, Sprague-Dawley , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Signal Transduction
9.
Biomed Pharmacother ; 144: 112280, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34628167

ABSTRACT

Metformin is currently used as a first-line drug to treat patients with type 2 diabetes. Previous studies have demonstrated that metformin has antioxidant properties and reduces neuroinflammation and hippocampal neuronal cell loss, which eventually improves memory. Methotrexate (MTX) is an antimetabolite chemotherapeutic agent reported to activate cognitive impairment found in many patients. Moreover, MTX negatively affects the spatial working memory, related to neurogenesis reduction in animal models. Therefore, the present study aimed to investigate the antioxidant effect of metformin on the reduction of memory and neurogenesis caused by MTX. Male Sprague-Dawley rats were divided into four groups: control, MTX, metformin, and MTX+metformin. MTX (75 mg/kg, i.v.) was administered on days 7 and 14. Rats were administered metformin (200 mg/kg, i.p.) for 14 days. Memory was determined using novel object location (NOL) and novel object recognition (NOR) tests. Furthermore, cell cycle arrest was quantified by p21 immunostaining. Levels of neuronal protein expression, scavenging enzymes activity, and malondialdehyde (MDA) level changes in the hippocampus and prefrontal cortex were investigated. Rats receiving only MTX showed memory impairment. Decreases in scavenging enzyme activity and BDNF, DCX, and Nrf2 protein expressions levels were detected in the MTX-treated rats. In addition, MTX significantly increased p21-positive cell numbers and MDA levels. However, these adverse MTX effects were counteracted by co-administration with metformin. These results demonstrate that metformin can improve memory impairments, increase BDNF, DCX and Nrf2 protein expressions and antioxidant capacities, and decrease MDA levels in MTX-treated rats.


Subject(s)
Behavior, Animal/drug effects , Hippocampus/drug effects , Memory Disorders/prevention & control , Memory/drug effects , Metformin/pharmacology , Neurogenesis/drug effects , Nootropic Agents/pharmacology , Oxidative Stress/drug effects , Prefrontal Cortex/drug effects , Animals , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Doublecortin Protein/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Male , Memory Disorders/chemically induced , Memory Disorders/metabolism , Memory Disorders/pathology , Methotrexate , NF-E2-Related Factor 2/metabolism , Open Field Test/drug effects , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Rats, Sprague-Dawley
10.
EMBO J ; 40(21): e107277, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34558085

ABSTRACT

The dorsal and ventral human telencephalons contain different neuronal subtypes, including glutamatergic, GABAergic, and cholinergic neurons, and how these neurons are generated during early development is not well understood. Using scRNA-seq and stringent validations, we reveal here a developmental roadmap for human telencephalic neurons. Both dorsal and ventral telencephalic radial glial cells (RGs) differentiate into neurons via dividing intermediate progenitor cells (IPCs_div) and early postmitotic neuroblasts (eNBs). The transcription factor ASCL1 plays a key role in promoting fate transition from RGs to IPCs_div in both regions. RGs from the regionalized neuroectoderm show heterogeneity, with restricted glutamatergic, GABAergic, and cholinergic differentiation potencies. During neurogenesis, IPCs_div gradually exit the cell cycle and branch into sister eNBs to generate distinct neuronal subtypes. Our findings highlight a general RGs-IPCs_div-eNBs developmental scheme for human telencephalic progenitors and support that the major neuronal fates of human telencephalon are predetermined during dorsoventral regionalization with neuronal diversity being further shaped during neurogenesis and neural circuit integration.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Lineage/genetics , Gene Expression Regulation, Developmental , Neurogenesis/genetics , Neurons/metabolism , Telencephalon/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle/genetics , Cell Differentiation , Choline/metabolism , Doublecortin Protein/genetics , Doublecortin Protein/metabolism , Fetus , Gene Ontology , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Glutamic Acid/metabolism , Humans , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Molecular Sequence Annotation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neurons/classification , Neurons/cytology , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Signal Transduction , Stathmin/genetics , Stathmin/metabolism , Telencephalon/cytology , Telencephalon/growth & development , Transcription Factors/genetics , Transcription Factors/metabolism , gamma-Aminobutyric Acid/metabolism
11.
PLoS One ; 16(8): e0256709, 2021.
Article in English | MEDLINE | ID: mdl-34464400

ABSTRACT

New neurons born in the adult brain undergo a critical period soon after migration to their site of incorporation. During this time, the behavior of the animal may influence the survival or culling of these cells. In the songbird song system, earlier work suggested that adult-born neurons may be retained in the song motor pathway nucleus HVC with respect to motor progression toward a target song during juvenile song learning, seasonal song restructuring, and experimentally manipulated song variability. However, it is not known whether the quality of song per se, without progressive improvement, may also influence new neuron survival. To test this idea, we experimentally altered song acoustic structure by unilateral denervation of the syrinx, causing a poor quality song. We found no effect of aberrant song on numbers of new neurons in HVC, suggesting that song quality does not influence new neuron culling in this region. However, aberrant song resulted in the loss of left-side dominance in new neurons in the auditory region caudomedial nidopallium (NCM), and a bilateral decrease in new neurons in the basal ganglia nucleus Area X. Thus new neuron culling may be influenced by behavioral feedback in accordance with the function of new neurons within that region. We propose that studying the effects of singing behaviors on new neurons across multiple brain regions that differentially subserve singing may give rise to general rules underlying the regulation of new neuron survival across taxa and brain regions more broadly.


Subject(s)
Geography , Neurogenesis , Vocal Cords/innervation , Vocalization, Animal/physiology , Aging/physiology , Animals , Doublecortin Protein/metabolism , Male , Neurons/physiology
12.
Cell Rep ; 36(1): 109324, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34233196

ABSTRACT

Adult neurogenesis in the dentate gyrus plays a role in adaptive brain functions such as memory formation. Adding new neurons to a specific locus of a neural circuit with functional needs is an efficient way to achieve such an adaptive function. However, it is unknown whether neurogenesis is linked to local functional demands potentially specified by the activity of neuronal circuits. By examining the distribution of neurogenesis and different types of neuronal activity in the dentate gyrus of freely moving adult rats, we find that neurogenesis is positionally associated with active excitatory neurons, some of which show place-cell activity, but is positionally dissociated from a type of interneuron with high-burst tendency. Our finding suggests that the behaviorally relevant activity of excitatory-inhibitory neuronal circuits can define a microenvironment stimulating/inhibiting neurogenesis. Such local regulation of neurogenesis may contribute to strategic recruitment of new neurons to modify functionally relevant neural circuits.


Subject(s)
Aging/physiology , Cellular Microenvironment , Dentate Gyrus/physiology , Nerve Net/physiology , Neural Inhibition/physiology , Neurogenesis , Action Potentials/physiology , Animals , Cell Proliferation , Doublecortin Protein/metabolism , Imaging, Three-Dimensional , Interneurons/physiology , Optogenetics , Place Cells/physiology , Rats, Long-Evans , Synapses/physiology
13.
Pharmacol Rep ; 73(4): 1164-1169, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33843023

ABSTRACT

BACKGROUND: Adult neurogenesis observed both in the subventricular zone (SVZ) and hippocampus may be regulated and modulated by several endogenous factors, xenobiotics and medications. Classical and atypical antipsychotic drugs are able to affect neuronal and glial cell proliferation in the rat brain. The main purpose of this structural study was to determine whether chronic chlorpromazine treatment affects adult neurogenesis in the canonical sites of the rat brain. At present, the clinical application of chlorpromazine is rather limited; however, it may still represent an important model in basic neuropharmacological and toxicological studies. METHODS: The number of neural progenitors and immature neurons was enumerated using immunofluorescent detection of Sox2, Musashi1 and doublecortin (DCX) expression within SVZ. RESULTS: Chlorpromazine has a depressive effect on the early phase of adult neurogenesis in the rat subventricular zone (SVZ), as the mean number of Sox-2 immunoexpressing cells decreased following treatment. CONCLUSION: Collectively, these results may suggest that long-term treatment with chlorpromazine may decrease neurogenic stem/progenitor cell formation in the rat SVZ and may affect rostral migratory stream formation.


Subject(s)
Chlorpromazine/adverse effects , Doublecortin Protein/metabolism , Lateral Ventricles/drug effects , Lateral Ventricles/metabolism , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Antipsychotic Agents/adverse effects , Cell Proliferation/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Male , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...