Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Cell Biosci ; 14(1): 92, 2024 Jul 14.
Article in English | MEDLINE | ID: mdl-39004750

ABSTRACT

BACKGROUND: N1-Methyladenosine (m1A) is an abundant modification of transcripts regulating mRNA structure and translation efficiency. However, the characteristics and biological functions of mRNA m1A modification in adult hippocampal neurogenesis remain enigmatic. RESULTS: We found that m1A demethylase Alkbh3 was dramatically enriched in neurons and neuronal genesis. Functionally, depletion of Alkbh3 in neural stem cells (NSCs) significantly decreased m1A modification, neuronal differentiation and proliferation coupling with increasing gliogenesis, whereas overexpressing Alkbh3 facilitated neuronal differentiation and proliferation. Mechanistically, the m1A demethylation of Mmp15 mRNA by Alkbh3 improved its RNA stability and translational efficacy, which promoted neurogenesis. Therapeutically, the silencing of Alkbh3 reduced hippocampal neurogenesis and impaired spatial memory in the adult mice. CONCLUSIONS: We reveal a novel function of m1A demethylation on Mmp15 mRNA in Alkbh3-mediated neurogenesis, which shed light on advancing Alkbh3 regulation of neurogenesis as a novel neurotherapeutic strategy.

2.
Cell Biosci ; 13(1): 183, 2023 Oct 01.
Article in English | MEDLINE | ID: mdl-37779199

ABSTRACT

BACKGROUND: N7-methylguanosine (m7G) is one of the most conserved modifications in nucleosides impacting mRNA export, splicing, and translation. However, the precise function and molecular mechanism of internal mRNA m7G methylation in adult hippocampal neurogenesis and neurogenesis-related Alzheimer's disease (AD) remain unknown. RESULTS: We profiled the dynamic Mettl1/Wdr4 expressions and m7G modification during neuronal differentiation of neural stem cells (NSCs) in vitro and in vivo. Adult hippocampal neurogenesis and its molecular mechanisms were examined by morphology, biochemical methods and biological sequencing. The translation efficiency of mRNA was detected by polysome profiling. The stability of Sptbn2 mRNA was constructed by RNA stability assay. APPswe/PS1ΔE9 (APP/PS1) double transgenic mice were used as model of AD. Morris water maze was used to detect the cognitive function. METHODS: We found that m7G methyltransferase complex Mettl1/Wdr4 as well as m7G was significantly elevated in neurons. Functionally, silencing Mettl1 in neural stem cells (NSCs) markedly decreased m7G modification, neuronal genesis and proliferation in addition to increasing gliogenesis, while forced expression of Mettl1 facilitated neuronal differentiation and proliferation. Mechanistically, the m7G modification of Sptbn2 mRNA by Mettl1 enhanced its stability and translation, which promoted neurogenesis. Importantly, genetic defciency of Mettl1 reduced hippocampal neurogenesis and spatial memory in the adult mice. Furthermore, Mettl1 overexpression in the hippocampus of APP/PS1 mice rescued neurogenesis and behavioral defects. CONCLUSION: Our findings unravel the pivotal role of internal mRNA m7G modification in Sptbn2-mediated neurogenesis, and highlight Mettl3 regulation of neurogenesis as a novel therapeutic target in AD treatment.

3.
Hum Vaccin Immunother ; 18(6): 2121568, 2022 Nov 30.
Article in English | MEDLINE | ID: mdl-36113067

ABSTRACT

Bacillus Calmette - Guerin (BCG) is an immune regulator that can enhance hippocampal synaptic plasticity in rats; however, it is unclear whether it can improve synaptic function in a mouse model with Alzheimer's disease (AD). We hypothesized that BCG plays a protective role in AD mice and investigated its effect on dendritic morphology. The results obtained show that BCG immunization significantly increases dendritic complexity, as indicated by the increased number of dendritic intersections and branch points, as well as the increase in the fractal dimension. Furthermore, the number of primary neurites and dendritic length also increased following BCG immunization, which increased the number of spines and promoted maturation. IFN-γ and IL-4 levels increased, while TNF-α levels decreased following BCG immunization; expression levels of p-JAK2, P-STAT3, SYN, and PSD-95 also increased. Therefore, this study demonstrates that BCG immunization in APP/PS1 mice mitigated hippocampal dendritic spine pathology, especially after the third round of immunization. This effect could possibly be attributed to; changes in dendritic arborization and spine morphology or increases in SYN and PSD-95 expression levels. It could also be related to mechanisms of BCG-induced increases in IFN-γ or IL-4/JAK2/STAT3 levels.


BCG immunization in a mouse model for Alzheimer's disease significantly increased dendritic complexity, as indicated by an increase in the number of dendritic intersections and branch points, as well as an increase in the fractal dimension of hippocampal CA1 neurons.


Subject(s)
Alzheimer Disease , BCG Vaccine , Dendrites , Animals , Mice , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Dendritic Spines/immunology , Dendritic Spines/metabolism , Dendritic Spines/pathology , Disease Models, Animal , Hippocampus/metabolism , Interleukin-4/metabolism , Mice, Transgenic , BCG Vaccine/therapeutic use , Dendrites/immunology , Dendrites/metabolism , Dendrites/pathology , Tumor Necrosis Factor-alpha/metabolism
4.
FASEB J ; 36(7): e22392, 2022 07.
Article in English | MEDLINE | ID: mdl-35716070

ABSTRACT

N6 -methyladenosine (m6 A) is the most abundant mRNA modification affecting diverse biological processes. However, the functions and precise mechanisms of m6 A signaling in adult hippocampal neurogenesis and neurogenesis-related depression remain largely enigmatic. We found that depletion of Mettl3 or Mettl14 in neural stem cells (NSCs) dramatically reduced m6 A abundance, proliferation, and neuronal genesis, coupled with enhanced glial differentiation. Conversely, overexpressing Mettl3 promoted proliferation and neuronal differentiation. Mechanistically, the m6 A modification of Lrp2 mRNA by Mettl3 enhanced its stability and translation efficiency relying on the reader protein Ythdc2, which in turn promoted neurogenesis. Importantly, mice lacking Mettl3 manifested reduced hippocampal neurogenesis, which could contribute to spatial memory decline, and depression-like behaviors. We found that these defective behaviors were notably reversed by Lrp2 overexpression. Moreover, Mettl3 overexpression in the hippocampus of depressive mice rescues behavioral defects. Our findings uncover the biological role of m6 A modification in Lrp2-mediated neurogenesis via m6 A-binding protein Ythdc2, and propose a rationale that targeting Mettl3-Ythdc2-Lrp2 axis regulation of neurogenesis might serve as a promising antidepressant strategy.


Subject(s)
Adenosine , Low Density Lipoprotein Receptor-Related Protein-2 , Methyltransferases , Neurogenesis , RNA Helicases , Adenosine/analogs & derivatives , Adenosine/metabolism , Animals , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Methyltransferases/metabolism , Mice , Neurogenesis/physiology , RNA Helicases/metabolism , RNA, Messenger/genetics
5.
Stem Cells ; 40(1): 59-73, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35511865

ABSTRACT

Increased neurogenesis elicits antidepressive-like effects. The antidiabetic drug metformin (Met) reportedly promotes hippocampal neurogenesis, which ameliorates spatial memory deficits and depression-like behaviors. However, the precise molecular mechanisms underpinning Met-induced neuronal differentiation of neural stem cells (NSCs) remain unclear. We showed that Met enhanced neuronal differentiation of NSCs via Gadd45g but not Gadd45a and Gadd45b. We further found that Gadd45g increased demethylation of neurogenic differentiation 1 promoter by regulating the activity of passive and active DNA demethylation enzymes through an adenylate-activated protein kinase -independent mechanism in Met-treated NSCs. Importantly, genetic deficiency of Gadd45g decreased hippocampal neurogenesis, which could contribute to spatial memory decline, and depression-like behaviors in the adult mice, whereas forced expression of Gadd45g alleviated the depressive-like behaviors. Our findings provide a model that Gadd45g-mediated DNA demethylation contributes to Met-induced neuronal genesis and its antidepressant-like effects and propose the concept that targeting Gadd45g regulation of neurogenesis might serve as a novel antidepressant strategy.


Subject(s)
Metformin , Neural Stem Cells , Animals , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacology , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , DNA Demethylation , Hippocampus/metabolism , Metformin/metabolism , Metformin/pharmacology , Mice , Neural Stem Cells/metabolism , Neurogenesis
6.
Pharmacol Res ; 179: 106235, 2022 05.
Article in English | MEDLINE | ID: mdl-35472635

ABSTRACT

Adult hippocampal neurogenesis (AHN) is heavily implicated in the pathogenesis of various neuropsychiatric disorders. The mangiferin (MGF), a bioactive compound of the mango, reportedly produces biological effects on a variety of neuropsychiatric disorders. However, the function and underlying mechanisms of MGF in regulating hippocampal neurogenesis remain unknown. Here we discovered that the transcriptome and methylome of MGF-induced neural stem cells (NSCs) are distinct from the control. RNA-seq analysis revealed that the diferentially expressed genes (DEGs) were signifcantly enriched in the PPARs. Furthermore, we found that MGF enhanced neuronal differentiation and proliferation of neural stem cells (NSCs) via PPARß but not PPARα and PPARγ. The combination of WGBS and RNA-seq analysis showed that the expression of some neurogenesis genes was negatively correlated with the DNA methylation level generally. We further found that PPARß increased demethylation of Mash1 promoter by modulating the expressions of active and passive DNA demethylation enzymes in MGF-treated NSCs. Importantly, genetic deficiency of PPARß decreased hippocampal neurogenesis in the adult mice, whereas the defective neurogenesis was notably rescued by Mash1 overexpression. Our findings uncover a model that PPARß-mediated DNA demethylation of Mash1 contributes to MGF-induced neuronal genesis, and advance the concept that targeting PPARß-TET1/DNMT3a-Mash1 axis regulation of neurogenesis might serve as a novel neurotherapeutic strategy.


Subject(s)
Neural Stem Cells , PPAR-beta , Animals , Mice , DNA Demethylation , Neurogenesis , PPAR-beta/genetics , PPAR-beta/metabolism , Xanthones
7.
Acta Neurol Scand ; 145(3): 273-278, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34779509

ABSTRACT

Microglia are a type of glial cells that play a key role in the repair of damage to the central nervous system (CNS). In the pathological condition of Alzheimer's disease (AD), ß-amyloid peptide and a variety of pro-inflammatory factors can activate microglia, resulting in the secretion of a variety of inflammatory factors and neurotoxins. This leads to neuronal damage and even apoptosis, thus triggering AD. In contrast, microglia can protect the CNS by phagocytizing Aß to slow down AD development. In this review, the roles of microglia in AD neuroinflammation and the scope of immunotherapy for AD are summarized to provide a theoretical basis for AD prevention and treatment.


Subject(s)
Alzheimer Disease , Alzheimer Disease/therapy , Amyloid beta-Peptides , Humans , Immunotherapy , Microglia , Neurons
8.
Front Neurosci ; 16: 1060111, 2022.
Article in English | MEDLINE | ID: mdl-36605552

ABSTRACT

Background and objective: Alzheimer's disease (AD) is the most common type of dementia, with its pathology like beta-amyloid and phosphorylated tau beginning several years before the clinical onset. The aim is to identify genetic risk factors associated with the onset of AD. Methods: We collected three microarray data of post-mortem brains of AD patients and the healthy from the GEO database and screened differentially expressed genes between AD and healthy control. GO/KEGG analysis was applied to identify AD-related pathways. Then we distinguished differential expressed genes between symptomatic and asymptomatic AD. Feature importance with logistic regression analysis is adopted to identify the most critical genes with symptomatic AD. Results: Data was collected from three datasets, including 184 AD patients and 132 healthy controls. We found 66 genes to be differently expressed between AD and the control. The pathway enriched in the process of exocytosis, synapse, and metabolism and identified 19 candidate genes, four of which (VSNL1, RTN1, FGF12, and ENC1) are vital. Conclusion: VSNL1, RTN1, FGF12, and ENC1 may be the essential genes that progress asymptomatic AD to symptomatic AD. Moreover, they may serve as genetic risk factors to identify high-risk individuals showing an earlier onset of AD.

9.
Cell Biosci ; 11(1): 29, 2021 Feb 03.
Article in English | MEDLINE | ID: mdl-33536056

ABSTRACT

BACKGROUND: The ketogenic diet (KD) has been recognized as a potentially effective therapy to treat neuropsychiatric diseases, including epilepsy. Previous studies have indicated that KD treatment elevates γ-Amino butyric acid (GABA) levels in both human and murine brains, which presumably contributes to the KD's anti-seizure effects. However, this has not been systematically investigated at the synaptic level, and the underlying molecular mechanisms remain to be elucidated. METHODS: Kainic acid (KA)-induced acute and chronic seizure models were utilized to examine the effects of KD treatment on seizure threshold and epileptogenesis. Synaptic activities in the hippocampus were recorded with the technique of electrophysiology. The effects of the KD on Neuregulin 1 (Nrg1) expression were assessed via RNA sequencing, real-time PCR and Western blotting. The obligatory role of Nrg1 in KD's effects on seizures was evaluated through disruption of Nrg1 signaling in mice by genetically deleting its receptor-ErbB4. RESULTS: We found that KD treatment suppressed seizures in both acute and chronic seizure models and enhanced presynaptic GABA release probability in the hippocampus. By screening molecular targets linked to GABAergic activity with transcriptome analysis, we identified that KD treatment dramatically increased the Nrg1 gene expression in the hippocampus. Disruption of Nrg1 signaling by genetically deleting its receptor-ErbB4 abolished KD's effects on GABAergic activity and seizures. CONCLUSION: Our findings suggest a critical role of Nrg1/ErbB4 signaling in mediating KD's effects on GABAergic activity and seizures, shedding light on developing new therapeutic interventions to seizure control.

10.
Cell Death Dis ; 11(10): 897, 2020 10 22.
Article in English | MEDLINE | ID: mdl-33093440

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disease characterized by Lewy body formation and progressive dopaminergic neuron death in the substantia nigra (SN). Genetic susceptibility is a strong risk factor for PD. Previously, a rare gain-of-function variant of GLUD2 glutamate dehydrogenase (T1492G) was reported to be associated with early onset in male PD patients; however, the function and underlying mechanism of this variant remains elusive. In the present study, we generated adeno-associated virus expressing GLUD2 and its mutant under the control of the glial fibrillary acidic protein promotor and injected the virus into the SN pars compacta of either untreated mice or 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD model mice. Our results demonstrate that GLUD2 mutation in MPTP-induced PD mice exacerbates movement deficits and nigral dopaminergic neuron death and reduces glutamate transporters expression and function. Using GC-Q-TOF/MS-based metabolomics, we determined that GLUD2 mutation damages mitochondrial function by decreasing succinate dehydrogenase activity to impede the tricarboxylic acid cycle in the SN of MPTP-induced PD mice. Accordingly, GLUD2 mutant mice had reduced energy metabolism and increased apoptosis, possibly due to downregulation of brain-derived neurotrophic factor/nuclear factor E2-related factor 2 signaling in in vitro and in vivo PD models. Collectively, our findings verify the function of GLUD2 in PD and unravel a mechanism by which a genetic variant in human GLUD2 may contribute to disease onset.


Subject(s)
Glutamate Dehydrogenase/genetics , Parkinson Disease/genetics , Succinate Dehydrogenase/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Amino Acid Transport System X-AG/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cell Death/drug effects , Cell Line, Tumor , Citric Acid Cycle/drug effects , Disease Models, Animal , Dopaminergic Neurons/metabolism , Genetic Predisposition to Disease , Glutamate Dehydrogenase/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/ultrastructure , Mutation , NF-E2-Related Factor 2/metabolism , Parkinson Disease/enzymology , Risk Factors , Substantia Nigra/drug effects , Substantia Nigra/metabolism
11.
Neuroscience ; 448: 94-106, 2020 11 10.
Article in English | MEDLINE | ID: mdl-32946950

ABSTRACT

Neuroinflammation contributes to neuronal death in cerebral ischemia. Urolithin A (UA), a gut microbial metabolite of ellagic acid, has emerged as a potential anti-inflammatory agent. However, its roles and precise mechanisms in stroke remain unknown. Here we found that UA treatment ameliorated infarction, neurological deficit scores, and spatial memory deficits after cerebral ischemia. Furthermore, UA significantly reduced neuron loss and promoted neurogenesis after ischemic stroke. We also found that UA attenuated apoptosis by regulating apoptotic-related proteins. Meanwhile, UA treatment inhibited glial activation via affecting inflammatory signaling pathways, specifically by enhancing cerebral AMPK and IκBa activation while decreasing the activation of Akt, P65NFκB, ERK, JNK, and P38MAPK. Our findings reveal a key role of UA against ischemic stroke through modulating apoptosis and neuroinflammation in mice.


Subject(s)
Brain Ischemia , Stroke , Animals , Apoptosis , Brain Ischemia/drug therapy , Coumarins/pharmacology , Mice , Signal Transduction , Stroke/drug therapy
12.
Brain Behav Immun ; 88: 748-762, 2020 08.
Article in English | MEDLINE | ID: mdl-32413556

ABSTRACT

Depression is a severe neuropsychiatric disorder, of which the underlying pathological mechanisms remain unclear. The ketogenic diet (KD) has been reported to exhibit preventative effects on depressive-like behaviors in rodents. However, the therapeutic effects of KD on depressive-like behaviors have not been illustrated thus far. Here, we found that KD treatment dramatically ameliorated depressive-like behaviors in both repeated social defeat stress (R-SDS) and lipopolysaccharide (LPS) models, indicating the potential therapeutic effects of KD on depression. Our electrophysiological studies further showed that neuronal excitability was increased in the lateral habenula (LHb) of mice exposed to R-SDS or LPS, which can be reversed in the presence of KD treatment. Moreover, R-SDS and LPS were also found to induce robust microglial inflammatory activation in the LHb. Importantly, these phenotypes were rescued in mice fed with KD. In addition, we found that the protein level of innate immune receptor Trem2 in the LHb was significantly decreased in depression models. Specific knockdown of Trem2 in LHb microglia induced depressive-like behaviors, increased neuronal excitability as well as robust microglial inflammatory activation. Altogether, we demonstrated the therapeutic effects of KD on depressive-like behaviors, which are probably mediated via the restoration of microglial inflammatory activation and neuronal excitability. Besides, we also proposed an unrecognized function of Trem2 in the LHb for depression. Our study sheds light on the pathogenesis of depression and thereby offers a potential therapeutic intervention.


Subject(s)
Diet, Ketogenic , Habenula , Neurons , Animals , Depression , Membrane Glycoproteins , Mice , Receptors, Immunologic
13.
Front Neurosci ; 14: 62, 2020.
Article in English | MEDLINE | ID: mdl-32116509

ABSTRACT

Fear extinction remains an unresolved challenge for behavioral exposure therapy in patients with post-traumatic stress disorder (PTSD). Previous reports have suggested that social support from either familiar or unfamiliar same-sex partners is beneficial to attenuating fear responses during fear extinction and renewal. Despite that, few studies have examined the effects of social support in advance on fear extinction and/or retrieval. It is also not clear whether social company by a receptive mating partner in advance facilitates fear extinction. In the present study, we address these questions by introducing a co-housing method, where fear-conditioned male mice are co-housed with or without a receptive mating partner prior to fear extinction. We found that while co-housing with an ovariectomized female mouse showed little effect on fear extinction or retrieval, social company by a receptive mating partner in advance dramatically facilitates fear extinction. In addition, the number of cFos-positive neurons in the basolateral amygdala (BLA) were also found to be reduced in male mice accompanied with receptive mating partner in response to fear extinction and retrieval, indicating diminished neuronal activation. Electrophysiological studies further showed that the excitability of excitatory neurons in BLA was decreased, which is probably due to the attenuated basal level of excitatory synaptic transmission. Together, our observations demonstrate an effect of social company by a receptive mating partner can facilitate fear extinction and afford a possible cellular mechanism.

14.
Mol Med Rep ; 21(3): 1172-1180, 2020 03.
Article in English | MEDLINE | ID: mdl-31922229

ABSTRACT

Alzheimer's disease (AD) is the most prevalent age­related neurodegenerative disorder. It is featured by the progressive accumulation of ß­amyloid (Aß) plaques and neurofibrillary tangles. This can eventually lead to a decrease of cholinergic neurons in the basal forebrain. Stem cell transplantation is an effective treatment for neurodegenerative diseases. Previous studies have revealed that different types of stem or progenitor cells can mitigate cognition impairment in different Alzheimer's disease mouse models. However, understanding the underlying mechanisms of neural stem cell (NSC) therapies for AD requires further investigation. In the present study, the effects and the underlying mechanisms of the treatment of AD by NSCs are reported. The latter were labelled with the enhanced green fluorescent protein (EGFP) prior to implantation into the bilateral hippocampus of an amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic (Tg) mouse model of AD. It was observed that the number of basal forebrain cholinergic neurons was restored and the expression of choline acetyltransferase (ChAT) protein was increased. Moreover, the levels of synaptophysin (SYP), postsynaptic density protein 95 (PSD­95) and microtubule­associated protein (MAP­2) were significantly increased in the hippocampus of NSC­treated AD mice. Notably, spatial learning and memory were both improved after transplantation of NSCs. In conclusion, the present study revealed that NSC transplantation improved learning and memory functions in an AD mouse model. This treatment allowed repairing of basal forebrain cholinergic neurons and increased the expression of the cognition­related proteins SYP, PSD­95 and MAP­2 in the hippocampus.


Subject(s)
Alzheimer Disease , Cholinergic Neurons , Learning , Memory , Neural Stem Cells , Presenilin-1 , Stem Cell Transplantation , Synapses , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Amyloid/genetics , Amyloid/metabolism , Animals , Cholinergic Neurons/metabolism , Cholinergic Neurons/pathology , Disease Models, Animal , Mice , Mice, Transgenic , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neural Stem Cells/transplantation , Presenilin-1/biosynthesis , Presenilin-1/genetics , Synapses/genetics , Synapses/metabolism , Synapses/pathology
15.
J Comp Neurol ; 528(3): 389-406, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31423581

ABSTRACT

Area prostriata (Pro) has been found to play important roles in the rapid processing of moving stimuli in the far peripheral visual field. However, the specific neural substrates responsible for these functions remain unknown. In this study, we first examined the location, extent, and topography of the rodent equivalent of the primate Pro based on cytoarchitecture and molecular markers. We then identified its intimate connections with the primary visual cortex (V1) using retrograde and anterograde tracers. Our main finding is that medial V1, which receives peripheral visual information, has strong reciprocal connections with the Pro in both rat and mouse while lateral V1 has significantly fewer such connections. The direct V1 inputs to the Pro provide at least one of the shortest pathways for visual information to reach the Pro, and may be crucial to the fast processing of unexpected stimuli in the peripheral visual field.


Subject(s)
Nerve Net/chemistry , Nerve Net/physiology , Visual Cortex/chemistry , Visual Cortex/physiology , Visual Pathways/chemistry , Visual Pathways/physiology , Animals , In Situ Hybridization/methods , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Visual Fields/physiology
16.
Brain Res Bull ; 149: 268-278, 2019 07.
Article in English | MEDLINE | ID: mdl-31051226

ABSTRACT

Previous study has demonstrated the neurobeneficial role of BCG and influenza vaccines. Based on this, our study concentrated on the synergistic effects on development of central nervous system by combined vaccination with BCG and influenza vaccines in rats. Our results displayed that pups combinedly vaccinated with BCG and influenza vaccines showed a significant enhance in spatial cognition, induction of LTP, hippocampal neurogenesis and morphology of dendritic spines compared with pups vaccinated with BCG solely. Furthermore, combined vaccination with BCG and influenza vaccines showed higher expression of BDNF, IGF-1, IL-4, IFN-γ and lower IL-1ß, TNF-α and IL-6 than BCG. Taken together, combined vaccination with BCG and influenza vaccines presented synergistic effects on spatial cognition and hippocampal plasticity in rats.


Subject(s)
BCG Vaccine/metabolism , Cognition/drug effects , Influenza Vaccines/metabolism , Animals , Animals, Newborn , BCG Vaccine/pharmacology , Brain/metabolism , Cytokines/drug effects , Cytokines/metabolism , Drug Synergism , Female , Hippocampus/drug effects , Hippocampus/metabolism , Influenza Vaccines/pharmacology , Male , Maze Learning/physiology , Neurogenesis/physiology , Neuronal Plasticity/drug effects , Rats , Rats, Sprague-Dawley , Spatial Navigation/drug effects , Temporal Lobe/metabolism , Vaccination
17.
J Neuroinflammation ; 16(1): 62, 2019 Mar 14.
Article in English | MEDLINE | ID: mdl-30871577

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by an abnormal accumulation of amyloid-ß (Aß) plaques, neuroinflammation, and impaired neurogenesis. Urolithin A (UA), a gut-microbial metabolite of ellagic acid, has been reported to exert anti-inflammatory effects in the brain. However, it is unknown whether UA exerts its properties of anti-inflammation and neuronal protection in the APPswe/PS1ΔE9 (APP/PS1) mouse model of AD. METHODS: Morris water maze was used to detect the cognitive function. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay was performed to detect neuronal apoptosis. Immunohistochemistry analyzed the response of glia, Aß deposition, and neurogenesis. The expression of inflammatory mediators were measured by enzyme-linked immunosorbent assay (ELISA) and quantitative real-time polymerase chain reaction (qRT-PCR). The modulating effects of UA on cell signaling pathways were assayed by Western blotting. RESULTS: We demonstrated that UA ameliorated cognitive impairment, prevented neuronal apoptosis, and enhanced neurogenesis in APP/PS1 mice. Furthermore, UA attenuated Aß deposition and peri-plaque microgliosis and astrocytosis in the cortex and hippocampus. We also found that UA affected critical cell signaling pathways, specifically by enhancing cerebral AMPK activation, decreasing the activation of P65NF-κB and P38MAPK, and suppressing Bace1 and APP degradation. CONCLUSIONS: Our results indicated that UA imparted cognitive protection by protecting neurons from death and triggering neurogenesis via anti-inflammatory signaling in APP/PS1 mice, suggesting that UA might be a promising therapeutic drug to treat AD.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Coumarins/therapeutic use , Cytokines/metabolism , Encephalitis/drug therapy , Gene Expression Regulation/drug effects , Memory Disorders/drug therapy , Alzheimer Disease/complications , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cytokines/genetics , Disease Models, Animal , Encephalitis/etiology , Female , Gene Expression Regulation/genetics , Gliosis/drug therapy , Gliosis/genetics , Maze Learning/drug effects , Memory Disorders/etiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Neurogenesis/genetics , Plaque, Amyloid/drug therapy , Plaque, Amyloid/etiology , Presenilin-1/genetics , Presenilin-1/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
18.
Brain Behav ; 9(4): e01246, 2019 04.
Article in English | MEDLINE | ID: mdl-30848079

ABSTRACT

INTRODUCTION: Beyond its application as an epilepsy therapy, the ketogenic diet (KD) has been considered a potential treatment for a variety of other neurological and metabolic disorders. However, whether KD promotes functional restoration by reducing the pathological processes underlying individual diseases or through some independent mechanisms is not clear. METHODS: In this study, we evaluated the effect of KD on a series of behaviors and synaptic functions of young adult naive mice. Wild-type C57BL/6J mice at age of 2-3 months were fed with control diet or KD for three months. Body weight and caloric intake were monitored throughout the experiments. We assessed behavioral performance with seizure induction, motor coordination and activity, anxiety level, spatial learning and memory, sociability, and depression. Synaptic transmission and long-term potentiation were also recorded. RESULTS: KD-fed mice performed equivalent to control-diet-fed mice in the behavioral tests and electrophysiological assays except exhibiting slower weight gain and increased seizure threshold. CONCLUSIONS: Our results contribute to the better understanding of effects of the KD on physiological behaviors and synaptic functions.


Subject(s)
Behavior, Animal/physiology , Brain/physiopathology , Diet, Ketogenic , Long-Term Potentiation/physiology , Seizures/physiopathology , Animals , Body Weight , Male , Mice , Mice, Inbred C57BL , Synaptic Transmission/physiology
19.
Brain Behav Immun ; 79: 159-173, 2019 07.
Article in English | MEDLINE | ID: mdl-30763768

ABSTRACT

Neuroinflammation, considered as a pathological hallmark of Alzheimer's disease (AD), has been demonstrated to affect hippocampal neurogenesis and cognitive function. Interleukin-6 (IL-6) is a proinflammatory cytokine known to modulate neurogenesis. However, the mechanisms are still largely unknown. Here, we reported that IL-6 suppressed neurogenesis via a JAK2/STAT3 signaling in neural stem cells (NSCs). Importantly, we found that NeuroD1 (Neurogenic differentiation 1) gene expression, which drives NSCs neurodifferentiation, was regulated by TET3 and DNMT1 in a JAK2/STAT3-dependent manner. We further found that JAK2/STAT3 inhibition enhanced demethylation of NeuroD1 regulatory elements in IL-6-treated cells, which is related to the significant upregulation of TET3 expression as well as the decreased expression of DNMT1. Furthermore, Inhibiting JAK2/STAT3 significantly rescued the memory deficits and hippocampal neurogenesis dysfunction in APP/PS1 mice. Our data suggest that JAK2/STAT3 signaling plays a vital role in suppressing neurogenesis of NSCs exposed to IL-6 at the epigenetic level, by regulating DNA methylation/demethylation.


Subject(s)
Janus Kinase 2/metabolism , Neurogenesis/physiology , STAT3 Transcription Factor/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Demethylation , DNA Methylation , Dioxygenases/genetics , Dioxygenases/metabolism , Hippocampus/metabolism , Humans , Interleukin-6/metabolism , Male , Mice , Mice, Transgenic , Neural Stem Cells/metabolism , Neurogenesis/immunology , Neuroimmunomodulation , Signal Transduction/immunology
20.
J Neuroinflammation ; 15(1): 268, 2018 Sep 18.
Article in English | MEDLINE | ID: mdl-30227858

ABSTRACT

BACKGROUND: Netrin-1 functions largely via combined receptors and downstream effectors. Evidence has shown that astrocytes express netrin-1 receptors, including DCC and UNC5H2. However, whether netrin-1 influences the function of astrocytes was previously unknown. METHODS: Lipopolysaccharide was used to stimulate the primary cultured astrocytes; interleukin release was used to track astrocyte activation. In vivo, shRNA and netrin-1 protein were injected in the mouse brain. Infarct volume, astrocyte activation, and interleukin release were used to observe the function of netrin-1 in neuroinflammation and brain injury after middle cerebral artery occlusion. RESULTS: Our results demonstrated that netrin-1 reduced lipopolysaccharide-induced interleukin-1ß and interleukin-12ß release in cultured astrocytes, and blockade of the UNC5H2 receptor with an antibody reversed this effect. Additionally, netrin-1 increased p-AKT and PPAR-γ expression in primary cultured astrocytes. In vivo studies showed that knockdown of netrin-1 increased astrocyte activation in the mouse brain after middle cerebral artery occlusion (p < 0.05). Moreover, injection of netrin-1 attenuated GFAP expression (netrin-1 0.27 ± 0.06 vs. BSA 0.62 ± 0.04, p < 0.001) and the release of interleukins and reduced infarct volume after brain ischemia (netrin-1 0.27 ± 0.06 vs. BSA 0.62 ± 0.04 mm3, p < 0.05). CONCLUSION: Our results indicate that netrin-1 is an important molecule in regulating astrocyte activation and neuroinflammation in cerebral ischemia and provides a potential target for ischemic stroke therapy.


Subject(s)
Astrocytes/drug effects , Brain/drug effects , Down-Regulation/drug effects , Infarction, Middle Cerebral Artery/physiopathology , Netrin-1/pharmacology , Animals , Cells, Cultured , Infarction, Middle Cerebral Artery/chemically induced , Interleukins/metabolism , Lipopolysaccharides , Mice , Netrin-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL