Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.668
Filter
1.
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
2.
ACS Appl Mater Interfaces ; 16(28): 36168-36193, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38954488

ABSTRACT

In the intricate landscape of Traumatic Brain Injury (TBI), the management of TBI remains a challenging task due to the extremely complex pathophysiological conditions and excessive release of reactive oxygen species (ROS) at the injury site and the limited regenerative capacities of the central nervous system (CNS). Existing pharmaceutical interventions are limited in their ability to efficiently cross the blood-brain barrier (BBB) and expeditiously target areas of brain inflammation. In response to these challenges herein, we designed novel mussel inspired polydopamine (PDA)-coated mesoporous silica nanoparticles (PDA-AMSNs) with excellent antioxidative ability to deliver a new potential therapeutic GSK-3ß inhibitor lead small molecule abbreviated as Neuro Chemical Modulator (NCM) at the TBI site using a neuroprotective peptide hydrogel (PANAP). PDA-AMSNs loaded with NCM (i.e., PDA-AMSN-D) into the matrix of PANAP were injected into the damaged area in an in vivo cryogenic brain injury model (CBI). This approach is specifically built while keeping the logic AND gate circuit as the primary focus. Where NCM and PDA-AMSNs act as two input signals and neurological functional recovery as a single output. Therapeutically, PDA-AMSN-D significantly decreased infarct volume, enhanced neurogenesis, rejuvenated BBB senescence, and accelerated neurological function recovery in a CBI.


Subject(s)
Antioxidants , Bivalvia , Brain Injuries, Traumatic , Indoles , Nanocomposites , Neurogenesis , Oxidative Stress , Polymers , Indoles/chemistry , Indoles/pharmacology , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/metabolism , Polymers/chemistry , Polymers/pharmacology , Animals , Oxidative Stress/drug effects , Antioxidants/chemistry , Antioxidants/pharmacology , Nanocomposites/chemistry , Bivalvia/chemistry , Neurogenesis/drug effects , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Silicon Dioxide/chemistry , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Male
3.
Sci Rep ; 14(1): 16082, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38992091

ABSTRACT

Regulation of physiological homeostasis, including energy balance, is thought to be modified by low levels of adult neurogenesis in the hypothalamus. Hormones such as oestradiol can influence both embryonic and adult hypothalamic neurogenic programs, demonstrating a sensitivity of hypothalamic neural progenitor cells to endogenous hormones. Previously we showed that gestational exposure to environmental levels of the xenoestrogen bisphenol A (BPA) changed neural progenitor cell behaviors in the embryo; however, we did not examine if these changes were permanent to affect adult neurogenesis. Here we investigated whether adult neuro- and/or gliogenesis were altered in mice prenatally exposed to BPA and placed on a high-fat diet challenge. Gestationally exposed adult female mice on a standard diet gained less weight than non-BPA controls, whereas gestationally exposed BPA females on a high-fat diet gained more weight than controls. Males exposed to gestational BPA showed no differences in weight gain relative to control males. Concomitantly, adult neurogenesis was increased in the VMH, DMH, and PVN of adult female mice exposed to BPA on standard diet, suggesting that disrupted adult neurogenesis might perturb normal energy balance regulation in females. These results add to growing evidence that low-dose BPA exposure in utero causes changes to adult hypothalamic function.


Subject(s)
Benzhydryl Compounds , Energy Metabolism , Homeostasis , Hypothalamus , Neurogenesis , Phenols , Prenatal Exposure Delayed Effects , Animals , Benzhydryl Compounds/toxicity , Female , Phenols/toxicity , Neurogenesis/drug effects , Pregnancy , Mice , Hypothalamus/drug effects , Hypothalamus/metabolism , Prenatal Exposure Delayed Effects/chemically induced , Homeostasis/drug effects , Energy Metabolism/drug effects , Male , Diet, High-Fat/adverse effects
4.
Neuropharmacology ; 257: 110058, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38960135

ABSTRACT

Postnatal hippocampal neurogenesis is essential for learning and memory. Hippocampal neural precursor cells (NPCs) can be induced to proliferate and differentiate into either glial cells or dentate granule cells. Notably, hippocampal neurogenesis decreases dramatically with age, partly due to a reduction in the NPC pool and a decrease in their proliferative activity. Alpha-melanocyte-stimulating hormone (α-MSH) improves learning, memory, neuronal survival and plasticity. Here, we used postnatally-isolated hippocampal NPCs from Wistar rat pups (male and female combined) to determine the role of the melanocortin analog [Nle4, D-Phe7]-α-MSH (NDP-MSH) in proliferation and fate acquisition of NPCs. Incubation of growth-factor deprived NPCs with 10 nM NDP-MSH for 6 days increased the proportion of Ki-67- and 5-bromo-2'-deoxyuridine (BrdU)-positive cells, compared to the control group, and these effects were blocked by the MC4R antagonist JKC-363. NDP-MSH also increased the proportion of glial fibrillar acidic protein (GFAP)/Ki-67, GFAP/sex-determining region Y-box2 (SOX2) and neuroepithelial stem cell protein (NESTIN)/Ki-67-double positive cells (type-1 and type-2 precursors). Finally, NDP-MSH induced peroxisome proliferator-activated receptor (PPAR)-γ protein expression, and co-incubation with the PPAR-γ inhibitor GW9662 prevented the effect of NDP-MSH on NPC proliferation and differentiation. Our results indicate that in vitro activation of MC4R in growth-factor-deprived postnatal hippocampal NPCs induces proliferation and promotes the relative expansion of the type-1 and type-2 NPC pool through a PPAR-γ-dependent mechanism. These results shed new light on the mechanisms underlying the beneficial effects of melanocortins in hippocampal plasticity and provide evidence linking the MC4R and PPAR-γ pathways in modulation of hippocampal NPC proliferation and differentiation.


Subject(s)
Cell Differentiation , Cell Proliferation , Hippocampus , Neural Stem Cells , Neurogenesis , Rats, Wistar , Receptor, Melanocortin, Type 4 , alpha-MSH , Animals , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Receptor, Melanocortin, Type 4/metabolism , alpha-MSH/pharmacology , alpha-MSH/analogs & derivatives , Female , Cell Differentiation/drug effects , Cell Differentiation/physiology , Male , Neurogenesis/drug effects , Neurogenesis/physiology , Rats , Cells, Cultured , SOXB1 Transcription Factors/metabolism , Animals, Newborn , Glial Fibrillary Acidic Protein/metabolism , PPAR gamma/metabolism
5.
Life Sci Alliance ; 7(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-38977310

ABSTRACT

Hippocampal seizures mimicking mesial temporal lobe epilepsy cause a profound disruption of the adult neurogenic niche in mice. Seizures provoke neural stem cells to switch to a reactive phenotype (reactive neural stem cells, React-NSCs) characterized by multibranched hypertrophic morphology, massive activation to enter mitosis, symmetric division, and final differentiation into reactive astrocytes. As a result, neurogenesis is chronically impaired. Here, using a mouse model of mesial temporal lobe epilepsy, we show that the epidermal growth factor receptor (EGFR) signaling pathway is key for the induction of React-NSCs and that its inhibition exerts a beneficial effect on the neurogenic niche. We show that during the initial days after the induction of seizures by a single intrahippocampal injection of kainic acid, a strong release of zinc and heparin-binding epidermal growth factor, both activators of the EGFR signaling pathway in neural stem cells, is produced. Administration of the EGFR inhibitor gefitinib, a chemotherapeutic in clinical phase IV, prevents the induction of React-NSCs and preserves neurogenesis.


Subject(s)
ErbB Receptors , Heparin-binding EGF-like Growth Factor , Hippocampus , Neural Stem Cells , Neurogenesis , Seizures , Signal Transduction , Animals , ErbB Receptors/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/drug effects , Hippocampus/metabolism , Mice , Heparin-binding EGF-like Growth Factor/metabolism , Seizures/metabolism , Neurogenesis/drug effects , Signal Transduction/drug effects , Male , Disease Models, Animal , Gefitinib/pharmacology , Epilepsy, Temporal Lobe/metabolism , Cell Differentiation/drug effects , Kainic Acid/pharmacology , Mice, Inbred C57BL
6.
Folia Biol (Praha) ; 70(1): 45-52, 2024.
Article in English | MEDLINE | ID: mdl-38830122

ABSTRACT

Effective treatment of patients with autism spectrum disorder (ASD) is still absent so far. Taurine exhibits therapeutic effects towards the autism-like behaviour in ASD model animals. Here, we determined the mechanism of taurine effect on hippocampal neurogenesis in genetically inbred BTBR T+ tf/J (BTBR) mice, a proposed model of ASD. In this ASD mouse model, we explored the effect of oral taurine supplementation on ASD-like behaviours in an open field test, elevated plus maze, marble burying test, self-grooming test, and three-chamber test. The mice were divided into four groups of normal controls (WT) and models (BTBR), who did or did not receive 6-week taurine supplementation in water (WT, WT+ Taurine, BTBR, and BTBR+Taurine). Neurogenesis-related effects were determined by Ki67 immunofluorescence staining. Western blot analysis was performed to detect the expression of phosphatase and tensin homologue deleted from chromosome 10 (PTEN)/mTOR/AKT pathway-associated proteins. Our results showed that taurine improved the autism-like behaviour, increased the proliferation of hippocampal cells, promoted PTEN expression, and reduced phosphorylation of mTOR and AKT in hippocampal tissue of the BTBR mice. In conclusion, taurine reduced the autism-like behaviour in partially inherited autism model mice, which may be associa-ted with improving the defective neural precursor cell proliferation and enhancing the PTEN-associated pathway in hippocampal tissue.


Subject(s)
Autistic Disorder , Hippocampus , Neurogenesis , PTEN Phosphohydrolase , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Taurine , Animals , Taurine/pharmacology , Hippocampus/metabolism , Hippocampus/drug effects , TOR Serine-Threonine Kinases/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Neurogenesis/drug effects , Autistic Disorder/metabolism , Autistic Disorder/drug therapy , Male , Behavior, Animal/drug effects , Mice , Disease Models, Animal , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/drug therapy , Cell Proliferation/drug effects
7.
Biomolecules ; 14(6)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38927091

ABSTRACT

BACKGROUND: Induced pluripotent stem cell (iPSC) based neuronal differentiation is valuable for studying neuropsychiatric disorders and pharmacological mechanisms at the cellular level. We aimed to examine the effects of typical and atypical antipsychotics on human iPSC-derived neural progenitor cells (NPCs). METHODS: Proliferation and neurite outgrowth were measured by live cell imaging, and gene expression levels related to neuronal identity were analyzed by RT-QPCR and immunocytochemistry during differentiation into hippocampal dentate gyrus granule cells following treatment of low- and high-dose antipsychotics (haloperidol, olanzapine, and risperidone). RESULTS: Antipsychotics did not modify the growth properties of NPCs after 3 days of treatment. However, the characteristics of neurite outgrowth changed significantly in response to haloperidol and olanzapine. After three weeks of differentiation, mRNA expression levels of the selected neuronal markers increased (except for MAP2), while antipsychotics caused only subtle changes. Additionally, we found no changes in MAP2 or GFAP protein expression levels as a result of antipsychotic treatment. CONCLUSIONS: Altogether, antipsychotic medications promoted neurogenesis in vitro by influencing neurite outgrowth rather than changing cell survival or gene expression. This study provides insights into the effects of antipsychotics on neuronal differentiation and highlights the importance of considering neurite outgrowth as a potential target of action.


Subject(s)
Antipsychotic Agents , Cell Differentiation , Haloperidol , Hippocampus , Induced Pluripotent Stem Cells , Neural Stem Cells , Neurogenesis , Olanzapine , Risperidone , Humans , Olanzapine/pharmacology , Risperidone/pharmacology , Neurogenesis/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Haloperidol/pharmacology , Antipsychotic Agents/pharmacology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Neuronal Outgrowth/drug effects
8.
PLoS One ; 19(6): e0305853, 2024.
Article in English | MEDLINE | ID: mdl-38913632

ABSTRACT

The intricate process of neuronal differentiation integrates multiple signals to induce transcriptional, morphological, and electrophysiological changes that reshape the properties of neural precursor cells during their maturation and migration process. An increasing number of neurotransmitters and biomolecules have been identified as molecular signals that trigger and guide this process. In this sense, taurine, a sulfur-containing, non-essential amino acid widely expressed in the mammal brain, modulates the neuronal differentiation process. In this study, we describe the effect of taurine acting via the ionotropic GABAA receptor and the metabotropic GABAB receptor on the neuronal differentiation and electrophysiological properties of precursor cells derived from the subventricular zone of the mouse brain. Taurine stimulates the number of neurites and favors the dendritic complexity of the neural precursor cells, accompanied by changes in the somatic input resistance and the strength of inward and outward membranal currents. At the pharmacological level, the blockade of GABAA receptors inhibits these effects, whereas the stimulation of GABAB receptors has no positive effects on the taurine-mediated differentiation process. Strikingly, the blockade of the GABAB receptor with CGP533737 stimulates neurite outgrowth, dendritic complexity, and membranal current kinetics of neural precursor cells. The effects of taurine on the differentiation process involve Ca2+ mobilization and the activation of intracellular signaling cascades since chelation of intracellular calcium with BAPTA-AM, and inhibition of the CaMKII, ERK1/2, and Src kinase inhibits the neurite outgrowth of neural precursor cells of the subventricular zone.


Subject(s)
Cell Differentiation , Lateral Ventricles , Neural Stem Cells , Receptors, GABA-A , Receptors, GABA-B , Animals , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Receptors, GABA-B/metabolism , Mice , Cell Differentiation/drug effects , Receptors, GABA-A/metabolism , Lateral Ventricles/cytology , Lateral Ventricles/metabolism , Taurine/pharmacology , Neurogenesis/drug effects , Calcium/metabolism
9.
Phytomedicine ; 131: 155802, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38852473

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is a serious neurodegenerative disease and brings a serious burden to society and families. Due to lack of effective drugs for the treatment of AD, it's urgent to develop new and effective drug for the treatment of AD. PURPOSE: The study aimed to investigate the potential of Zexieyin formula (ZXYF), a Chinese medicine formula, for the treatment of AD and its potential mechanism of action. METHODS: We used chronic scopolamine (SCOP) induction mice model and APP/PS1 mice to reveal and confirm ZXYF for the treatment of AD with donepezil (DON) as a positive reference. The learning and memory function were detected by morris water maze test (MWM) and y-maze test. Moreover, western blot and immunofluorescence were used to detect the molecular mechanism of ZXYF for the alleviation of AD in hippocampus. Lastly, pharmacological technology was applied to evaluate AMPA receptor involved in the role of ZXYF in the treatment of AD. RESULTS: The results showed that ZXYF could improve memory and learning deficits both in two AD models including scopolamine (SCOP)-induced mice model and APP/PS1mice. Moreover, ZXYF or not DON increased expressions of BrdU/DCX and Ki67 positive cells in dentate gyrus (DG), up-regulated the levels of AMPA subunit type (GluA1) and PKA in hippocampus in SCOP-induced mice model, although ZXYF and DON activated CaMKII, CaMKII-phosphorylation, CREB, CREB-phosphorylation and PSD95 in hippocampus in SCOP-induced mice model. ZXYF also activated CaMKII, CaMKII-phosphorylation and GluA1 in HT22 cells. Furthermore, transient inhibiting AMPA receptor was capable of blocking the effects of ZXYF to treat AD in MWM and suppressing the number of BrdU/DCX positive cells increased by ZXYF in DG in SCOP-induced mice model, but had no effect on the alteration of Ki67 positive cells. CONCLUSION: ZXYF had the therapeutic effects on AD-treatment, which activated CaMKII to promote AMPA receptor (GluA1) and subsequently up-regulated PKA/CREB signaling to facilitate neurogenesis to achieve enhanced postsynaptic protein.


Subject(s)
Alzheimer Disease , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Disease Models, Animal , Drugs, Chinese Herbal , Hippocampus , Neurogenesis , Neuronal Plasticity , Receptors, AMPA , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/chemically induced , Receptors, AMPA/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Drugs, Chinese Herbal/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Neurogenesis/drug effects , Mice , Male , Neuronal Plasticity/drug effects , Scopolamine , Mice, Transgenic , Maze Learning/drug effects , Donepezil/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Memory/drug effects , Mice, Inbred C57BL
10.
Cell Biol Toxicol ; 40(1): 41, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38833095

ABSTRACT

Hippocampal neurons maintain the ability of proliferation throughout life to support neurogenesis. Deoxynivalenol (DON) is a mycotoxin that exhibits brain toxicity, yet whether and how DON affects hippocampal neurogenesis remains unknown. Here, we use mouse hippocampal neuron cells (HT-22) as a model to illustrate the effects of DON on neuron proliferation and to explore underlying mechanisms. DON exposure significantly inhibits the proliferation of HT-22 cells, which is associated with an up-regulation of cell cycle inhibitor p21 at both mRNA and protein levels. Global and site-specific m6A methylation levels on the 3'UTR of p21 mRNA are significantly increased in response to DON treatment, whereas inhibition of m6A hypermethylation significantly alleviates DON-induced cell cycle arrest. Further mechanistic studies indicate that the m6A readers YTHDF1 and IGF2BP1 are responsible for m6A-mediated increase in p21 mRNA stability. Meanwhile, 3'UTR of E3 ubiquitin ligase TRIM21 mRNA is also m6A hypermethylated, and another m6A reader YTHDF2 binds to the m6A sites, leading to decreased TRIM21 mRNA stability. Consequently, TRIM21 suppression impairs ubiquitin-mediated p21 protein degradation. Taken together, m6A-mediated upregulation of p21, at both post-transcriptional and post-translational levels, contributes to DON-induced inhibition of hippocampal neuron proliferation. These results may provide new insights for epigenetic therapy of neurodegenerative diseases.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21 , Hippocampus , Neurons , Trichothecenes , Up-Regulation , Animals , Trichothecenes/toxicity , Trichothecenes/pharmacology , Hippocampus/metabolism , Hippocampus/drug effects , Hippocampus/cytology , Mice , Neurons/drug effects , Neurons/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Up-Regulation/drug effects , Cell Proliferation/drug effects , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Cell Line , 3' Untranslated Regions/genetics , Neurogenesis/drug effects , RNA, Messenger/metabolism , RNA, Messenger/genetics , RNA Stability/drug effects , Cell Cycle Checkpoints/drug effects , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Methylation/drug effects
11.
Nutrients ; 16(12)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38931326

ABSTRACT

Taurine (2-aminoethanesulfonic acid) is a non-protein ß-amino acid essential for cellular homeostasis, with antioxidant, anti-inflammatory, and cytoprotective properties that are crucial for life maintenance. This study aimed to evaluate the effects of taurine administration on hippocampal neurogenesis, neuronal preservation, or reverse damage in rats exposed to forced ethanol consumption in an animal model. Wistar rats were treated with ethanol (EtOH) for a 28-day period (5% in the 1st week, 10% in the 2nd week, and 20% in the 3rd and 4th weeks). Two taurine treatment protocols (300 mg/kg i.p.) were implemented: one during ethanol consumption to analyze neuroprotection, and another after ethanol consumption to assess the reversal of ethanol-induced damage. Overall, the results demonstrated that taurine treatment was effective in protecting against deficits induced by ethanol consumption in the dentate gyrus. The EtOH+TAU group showed a significant increase in cell proliferation (145.8%) and cell survival (54.0%) compared to the EtOH+Sal group. The results also indicated similar effects regarding the reversal of ethanol-induced damage 28 days after the cessation of ethanol consumption. The EtOH+TAU group exhibited a significant increase (41.3%) in the number of DCX-immunoreactive cells compared to the EtOH+Sal group. However, this amino acid did not induce neurogenesis in the tissues of healthy rats, implying that its activity may be contingent upon post-injury stimuli.


Subject(s)
Doublecortin Protein , Ethanol , Hippocampus , Neurogenesis , Neuroprotective Agents , Rats, Wistar , Taurine , Animals , Taurine/pharmacology , Neurogenesis/drug effects , Male , Neuroprotective Agents/pharmacology , Rats , Hippocampus/drug effects , Cell Proliferation/drug effects , Dentate Gyrus/drug effects , Neurons/drug effects , Cell Survival/drug effects , Disease Models, Animal
12.
CNS Neurosci Ther ; 30(6): e14800, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38887162

ABSTRACT

BACKGROUND: Impaired mitochondrial dynamics have been identified as a significant contributing factor to reduced neurogenesis under pathological conditions. However, the relationship among mitochondrial dynamics, neurogenesis, and spatial memory during normal development remains unclear. This study aims to elucidate the role of mitophagy in spatial memory mediated by neurogenesis during development. METHODS: Adolescent and adult male mice were used to assess spatial memory performance. Immunofluorescence staining was employed to evaluate levels of neurogenesis, and mitochondrial dynamics were assessed through western blotting and transmission electron microscopy. Pharmacological interventions further validated the causal relationship among mitophagy, neurogenesis, and behavioral performance during development. RESULTS: The study revealed differences in spatial memory between adolescent and adult mice. Diminished neurogenesis, accompanied by reduced mitophagy, was observed in the hippocampus of adult mice compared to adolescent subjects. Pharmacological induction of mitophagy in adult mice with UMI-77 resulted in enhanced neurogenesis and prolonged spatial memory retention. Conversely, inhibition of mitophagy with Mdivi-1 in adolescent mice led to reduced hippocampal neurogenesis and impaired spatial memory. CONCLUSION: The observed decline in spatial memory in adult mice is associated with decreased mitophagy, which affects neurogenesis in the dentate gyrus. This underscores the therapeutic potential of enhancing mitophagy to counteract age- or disease-related cognitive decline.


Subject(s)
Hippocampus , Mitophagy , Neurogenesis , Spatial Memory , Animals , Neurogenesis/physiology , Neurogenesis/drug effects , Mitophagy/physiology , Mitophagy/drug effects , Spatial Memory/physiology , Male , Mice , Mice, Inbred C57BL , Mitochondrial Dynamics/physiology , Quinazolinones
13.
J Mater Chem B ; 12(25): 6221-6241, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38835196

ABSTRACT

Traumatic injuries, neurodegenerative diseases and oxidative stress serve as the early biomarkers for neuronal damage and impede angiogenesis and subsequently neuronal growth. Considering this, the present work aimed to develop a poly(N-acryloylglycine)-co-(acrylamide)-co-(N-acryloylglutamate) hydrogel [p(NAG-Ac-NAE)] with angiogenesis/neurogenesis properties. As constituents of this polymer modulate their vital role in biological functions, inhibitory neurotransmitter glycine regulates neuronal homeostasis, and glutamatergic signalling regulates angiogenesis. The p(NAG-Ac-NAE) hydrogel is a highly branched, biodegradable and pH-responsive polymer with a very high swelling behavior of 6188%. The mechanical stability (G', 2.3-2.7 kPa) of this polymeric hydrogel is commendable in the differentiation of mature neurons. This hydrogel is biocompatible (as tested in HUVEC cells) and helps to proliferate PC12 cells (152.7 ± 13.7%), whereas it is cytotoxic towards aggressive cancers such as glioblastoma (LN229 cells) and triple negative breast cancer (TNBC; MDA-MB-231 cells) and helps to maintain the healthy cytoskeleton framework structure of primary cortical neurons by facilitating the elongation of the axonal pathway. Furthermore, FACS results revealed that the synthesized hydrogel potentiates neurogenesis by inducing the cell cycle (G0/G1) and arresting the sub-G1 phase by limiting apoptosis. Additionally, RT-PCR results revealed that this hydrogel induced an increased level of HIF-1α expression, providing preconditioning effects towards neuronal cells under oxidative stress by scavenging ROS and initiating neurogenic and angiogenic signalling. This hydrogel further exhibits more pro-angiogenic activities by increasing the expression of VEGF isoforms compared to previously reported hydrogels. In conclusion, the newly synthesized p(NAG-Ac-NAE) hydrogel can be one of the potential neuroregenerative materials for vasculogenesis-assisted neurogenic applications and paramount for the management of neurodegenerative diseases.


Subject(s)
Hydrogels , Oxidative Stress , Oxidative Stress/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Hydrogels/chemical synthesis , Humans , Animals , Rats , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Biocompatible Materials/chemical synthesis , Neurogenesis/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , PC12 Cells , Neovascularization, Physiologic/drug effects , Cell Proliferation/drug effects , Polymers/chemistry , Polymers/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis
14.
Int J Mol Sci ; 25(12)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38928164

ABSTRACT

Neurogenesis is the process by which new brain cells are formed. This crucial event emerges during embryonic life and proceeds in adulthood, and it could be influenced by environmental pollution. Non-combustion-derived magnetite represents a portion of the coarse particulate matter (PM) contributing to air and water pollution in urban settings. Studies on humans have reported that magnetite and other iron oxides have significant damaging effects at a central level, where these particles accumulate and promote oxidative stress. Similarly, magnetite nanoparticles can cross the placenta and damage the embryo brain during development, but the impact on neurogenesis is still unknown. Furthermore, an abnormal Fe cation concentration in cells and tissues might promote reactive oxygen species (ROS) generation and has been associated with multiple neurodegenerative conditions. In the present study, we used zebrafish as an in vivo system to analyze the specific effects of magnetite on embryonic neurogenesis. First, we characterized magnetite using mineralogical and spectroscopic analyses. Embryos treated with magnetite at sub-lethal concentrations showed a dose-response increase in ROS in the brain, which was accompanied by a massive decrease in antioxidant genes (sod2, cat, gsr, and nrf2). In addition, a higher number of apoptotic cells was observed in embryos treated with magnetite. Next, interestingly, embryos exposed to magnetite displayed a decrease in neural staminal progenitors (nestin, sox2, and pcna markers) and a neuronal marker (elavl3). Finally, we observed significative increases in apoeb (specific microglia marker) and interleukin-1b (il1b), confirming a status of inflammation in the brain embryos treated with magnetite. Our study represents the very first in vivo evidence concerning the effects of magnetite on brain development.


Subject(s)
Embryo, Nonmammalian , Ferrosoferric Oxide , Neurogenesis , Zebrafish , Animals , Zebrafish/embryology , Neurogenesis/drug effects , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Reactive Oxygen Species/metabolism , Oxidative Stress/drug effects , Brain/metabolism , Brain/drug effects , Brain/embryology , Apoptosis/drug effects , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/toxicity
15.
Int J Mol Sci ; 25(12)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38928248

ABSTRACT

Fluoxetine, a commonly prescribed medication for depression, has been studied in Alzheimer's disease (AD) patients for its effectiveness on cognitive symptoms. The aim of this systematic review is to investigate the therapeutic potential of fluoxetine in cognitive decline in AD, focusing on its anti-degenerative mechanisms of action and clinical implications. According to PRISMA, we searched MEDLINE, up to 1 April 2024, for animal and human studies examining the efficacy of fluoxetine with regard to the recovery of cognitive function in AD. Methodological quality was evaluated using the ARRIVE tool for animal AD studies and the Cochrane tool for clinical trials. In total, 22 studies were analyzed (19 animal AD studies and 3 clinical studies). Fluoxetine promoted neurogenesis and enhanced synaptic plasticity in preclinical models of AD, through a decrease in Aß pathology and increase in BDNF, by activating diverse pathways (such as the DAF-16-mediated, TGF-beta1, ILK-AKT-GSK3beta, and CREB/p-CREB/BDNF). In addition, fluoxetine has anti-inflammatory properties/antioxidant effects via targeting antioxidant Nrf2/HO-1 and hindering TLR4/NLRP3 inflammasome. Only three clinical studies showed that fluoxetine ameliorated the cognitive performance of people with AD; however, several methodological issues limited the generalizability of these results. Overall, the high-quality preclinical evidence suggests that fluoxetine may have neuroprotective, antioxidant, and anti-inflammatory effects in AD animal models. While more high-quality clinical research is needed to fully understand the mechanisms underlying these effects, fluoxetine is a promising potential treatment for AD patients. If future clinical trials confirm its anti-degenerative and neuroprotective effects, fluoxetine could offer a new therapeutic approach for slowing down the progression of AD.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Fluoxetine , Fluoxetine/therapeutic use , Fluoxetine/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Humans , Animals , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Disease Models, Animal , Neurogenesis/drug effects , Neuronal Plasticity/drug effects
16.
Int J Nanomedicine ; 19: 6035-6055, 2024.
Article in English | MEDLINE | ID: mdl-38911505

ABSTRACT

Background: Autism Spectrum Disorder (ASD) is a neurodevelopmental condition that affects social interaction and communication and can cause stereotypic behavior. Fullerenols, a type of carbon nanomaterial known for its neuroprotective properties, have not yet been studied for their potential in treating ASD. We aimed to investigate its role in improving autistic behaviors in BTBR T+Itpr3tf/J (BTBR) mice and its underlying mechanism, which could provide reliable clues for future ASD treatments. Methods: Our research involved treating C57BL/6J (C57) and BTBR mice with either 0.9% NaCl or fullerenols (10 mg/kg) daily for one week at seven weeks of age. We then conducted ASD-related behavioral tests in the eighth week and used RNA-seq to screen for vital pathways in the mouse hippocampus. Additionally, we used real-time quantitative PCR (RT-qPCR) to verify related pathway genes and evaluated the number of stem cells in the hippocampal dentate gyrus (DG) by Immunofluorescence staining. Results: Our findings revealed that fullerenols treatment significantly improved the related ASD-like behaviors of BTBR mice, manifested by enhanced social ability and improved cognitive deficits. Immunofluorescence results showed that fullerenols treatment increased the number of DCX+ and SOX2+/GFAP+ cells in the DG region of BTBR mice, indicating an expanded neural progenitor cell (NPC) pool of BTBR mice. RNA-seq analysis of the mouse hippocampus showed that VEGFA was involved in the rescued hippocampal neurogenesis by fullerenols treatment. Conclusion: In conclusion, our findings suggest that fullerenols treatment improves ASD-like behavior in BTBR mice by upregulating VEGFA, making nanoparticle- fullerenols a promising drug for ASD treatment.


Subject(s)
Autism Spectrum Disorder , Cognitive Dysfunction , Disease Models, Animal , Doublecortin Protein , Fullerenes , Mice, Inbred C57BL , Animals , Mice , Fullerenes/pharmacology , Fullerenes/chemistry , Autism Spectrum Disorder/drug therapy , Cognitive Dysfunction/drug therapy , Male , Social Behavior , Behavior, Animal/drug effects , Hippocampus/drug effects , Vascular Endothelial Growth Factor A/genetics , Neuroprotective Agents/pharmacology , Neurogenesis/drug effects , Autistic Disorder/drug therapy
17.
Biosci Rep ; 44(5)2024 May 29.
Article in English | MEDLINE | ID: mdl-38700092

ABSTRACT

Pre-eclampsia (PE) is a hypertensive disorder of pregnancy which is associated with increased risk of neurodevelopmental disorders in exposed offspring. The pathophysiological mechanisms mediating this relationship are currently unknown, and one potential candidate is the anti-angiogenic factor soluble Fms-like tyrosine kinase 1 (sFlt-1), which is highly elevated in PE. While sFlt-1 can impair angiogenesis via inhibition of VEGFA signalling, it is unclear whether it can directly affect neuronal development independently of its effects on the vasculature. To test this hypothesis, the current study differentiated the human neural progenitor cell (NPC) line ReNcell® VM into a mixed culture of mature neurons and glia, and exposed them to sFlt-1 during development. Outcomes measured were neurite growth, cytotoxicity, mRNA expression of nestin, MBP, GFAP, and ßIII-tubulin, and neurosphere differentiation. sFlt-1 induced a significant reduction in neurite growth and this effect was timing- and dose-dependent up to 100 ng/ml, with no effect on cytotoxicity. sFlt-1 (100 ng/ml) also reduced ßIII-tubulin mRNA and neuronal differentiation of neurospheres. Undifferentiated NPCs and mature neurons/glia expressed VEGFA and VEGFR-2, required for endogenous autocrine and paracrine VEGFA signalling, while sFlt-1 treatment prevented the neurogenic effects of exogenous VEGFA. Overall, these data provide the first experimental evidence for a direct effect of sFlt-1 on neurite growth and neuronal differentiation in human neurons through inhibition of VEGFA signalling, clarifying our understanding of the potential role of sFlt-1 as a mechanism by which PE can affect neuronal development.


Subject(s)
Neurites , Neurogenesis , Neurons , Vascular Endothelial Growth Factor Receptor-1 , Female , Humans , Pregnancy , Cell Line, Tumor , Neural Stem Cells/metabolism , Neural Stem Cells/drug effects , Neurites/metabolism , Neurites/drug effects , Neurogenesis/drug effects , Neurons/metabolism , Neurons/drug effects , Neurons/cytology , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics
18.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732109

ABSTRACT

Adipose-derived mesenchymal stem cells (ASCs) are adult multipotent stem cells, able to differentiate toward neural elements other than cells of mesodermal lineage. The aim of this research was to test ASC neural differentiation using melatonin combined with conditioned media (CM) from glial cells. Isolated from the lipoaspirate of healthy donors, ASCs were expanded in a basal growth medium before undergoing neural differentiation procedures. For this purpose, CM obtained from olfactory ensheathing cells and from Schwann cells were used. In some samples, 1 µM of melatonin was added. After 1 and 7 days of culture, cells were studied using immunocytochemistry and flow cytometry to evaluate neural marker expression (Nestin, MAP2, Synapsin I, GFAP) under different conditions. The results confirmed that a successful neural differentiation was achieved by glial CM, whereas the addition of melatonin alone did not induce appreciable changes. When melatonin was combined with CM, ASC neural differentiation was enhanced, as demonstrated by a further improvement of neuronal marker expression, whereas glial differentiation was attenuated. A dynamic modulation was also observed, testing the expression of melatonin receptors. In conclusion, our data suggest that melatonin's neurogenic differentiation ability can be usefully exploited to obtain neuronal-like differentiated ASCs for potential therapeutic strategies.


Subject(s)
Cell Differentiation , Melatonin , Mesenchymal Stem Cells , Melatonin/pharmacology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Humans , Cell Differentiation/drug effects , Cells, Cultured , Adipose Tissue/cytology , Neurons/cytology , Neurons/metabolism , Neurons/drug effects , Culture Media, Conditioned/pharmacology , Schwann Cells/cytology , Schwann Cells/metabolism , Schwann Cells/drug effects , Neurogenesis/drug effects , Adult , Nestin/metabolism , Nestin/genetics , Glial Fibrillary Acidic Protein/metabolism , Neuroglia/drug effects , Neuroglia/cytology , Neuroglia/metabolism , Synapsins/metabolism
19.
J Ethnopharmacol ; 331: 118332, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38735421

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Citri Reticulata Pericarpium Viride (also known Qing-Pi or QP) is a plant in the Rutaceae family, QP is a traditional Qi-regulating medicine in Chinese medicine that is compatible with other Chinese medicine components and has extensive clinical practice in treating anxiety and depression. Reports on the pharmacological effects of QP have demonstrated its neuroprotective effects and antioxidant capacities. Numerous pharmacological benefits of QP are attributed to its antioxidant abilities. Anxiety disorders are a broadly defined category of mental illnesses. Oxidative stress and an imbalance in the antioxidant defense system are typical pathological features of these disorders. AIM OF THE STUDY: The aim of this study was to evaluate the effects of QP essential oil on anxiety using animal models and investigate the underlying neurobiological mechanisms. MATERIALS AND METHODS: This study aimed to develop an animal model of anxiety using chronic restraint stress and investigate the effects of inhalation of Citri Reticulata Pericarpium Viride essential oil on anxiety-like behavior, olfactory function, and olfactory bulb neurogenesis in mice with anxiety. RESULTS: The results showed that long-term chronic restraint stimulation caused a decrease in olfactory function, significant anxiety-like behavior, and a notable reduction in the number of neurons in the olfactory bulb. However, inhalation of Citri Reticulata Pericarpium Viride essential oil reversed these effects, improving the olfactory function, neuro-stimulating effect, alleviating anxiety-like behavior, and regulating theta (4-12Hz) oscillation in the hippocampus DG area. These effects were associated with changes in the expression levels of glutamate receptor NMDAR and NeuN in olfactory bulb. CONCLUSIONS: The study revealed that mice with anxiety induced by chronic restraint stress exhibited significant olfactory dysfunction, providing strong evidence for the causal relationship between anxiety disorders and olfactory dysfunction. Moreover, QP essential oil has the potential to be developed as a therapeutic drug for anxiety disorders, in addition to its role as a complementary anxiolytic.


Subject(s)
Anti-Anxiety Agents , Anxiety , Oils, Volatile , Olfactory Bulb , Receptors, N-Methyl-D-Aspartate , Animals , Oils, Volatile/pharmacology , Oils, Volatile/isolation & purification , Male , Anxiety/drug therapy , Mice , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Anti-Anxiety Agents/isolation & purification , Receptors, N-Methyl-D-Aspartate/metabolism , Behavior, Animal/drug effects , Glutamic Acid/metabolism , Neurogenesis/drug effects , Disease Models, Animal , Stress, Psychological/drug therapy
20.
Neuron ; 112(9): 1373-1375, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38697018

ABSTRACT

Maternal well-being is important for the development of the fetus, with a key influence on its nervous system. In this issue of Neuron, Krontira et al.1 implicate glucocorticoids, the stress hormones, in the regulation of neural stem cell identity and proliferation, with long-lasting consequences on brain architecture and educational attainment.


Subject(s)
Glucocorticoids , Neurogenesis , Humans , Glucocorticoids/pharmacology , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/drug effects , Neurons/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/cytology , Neural Stem Cells/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL