Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Stem Cells ; 40(1): 59-73, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-35511865

RESUMEN

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.


Asunto(s)
Metformina , Células-Madre Neurales , Animales , Antidepresivos/metabolismo , Antidepresivos/farmacología , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Desmetilación del ADN , Hipocampo/metabolismo , Metformina/metabolismo , Metformina/farmacología , Ratones , Células-Madre Neurales/metabolismo , Neurogénesis
2.
FASEB J ; 36(7): e22392, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35716070

RESUMEN

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.


Asunto(s)
Adenosina , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad , Metiltransferasas , Neurogénesis , ARN Helicasas , Adenosina/análogos & derivados , Adenosina/metabolismo , Animales , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Metiltransferasas/metabolismo , Ratones , Neurogénesis/fisiología , ARN Helicasas/metabolismo , ARN Mensajero/genética
3.
Pharmacol Res ; 179: 106235, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35472635

RESUMEN

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.


Asunto(s)
Células-Madre Neurales , PPAR-beta , Animales , Ratones , Desmetilación del ADN , Neurogénesis , PPAR-beta/genética , PPAR-beta/metabolismo , Xantonas
4.
Acta Neurol Scand ; 145(3): 273-278, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34779509

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides , Humanos , Inmunoterapia , Microglía , Neuronas
5.
Brain Behav Immun ; 88: 748-762, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32413556

RESUMEN

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.


Asunto(s)
Dieta Cetogénica , Habénula , Neuronas , Animales , Depresión , Glicoproteínas de Membrana , Ratones , Receptores Inmunológicos
6.
J Neurosci ; 38(44): 9600-9613, 2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-30228230

RESUMEN

Fear learning and memory are vital for livings to survive, dysfunctions in which have been implicated in various neuropsychiatric disorders. Appropriate neuronal activation in amygdala is critical for fear memory. However, the underlying regulatory mechanisms are not well understood. Here we report that Neogenin, a DCC (deleted in colorectal cancer) family receptor, which plays important roles in axon navigation and adult neurogenesis, is enriched in excitatory neurons in BLA (Basolateral amygdala). Fear memory is impaired in male Neogenin mutant mice. The number of cFos+ neurons in response to tone-cued fear training was reduced in mutant mice, indicating aberrant neuronal activation in the absence of Neogenin. Electrophysiological studies show that Neogenin mutation reduced the cortical afferent input to BLA pyramidal neurons and compromised both induction and maintenance of Long-Term Potentiation evoked by stimulating cortical afferent, suggesting a role of Neogenin in synaptic plasticity. Concomitantly, there was a reduction in spine density and in frequency of miniature excitatory postsynaptic currents (mEPSCs), but not miniature inhibitory postsynaptic currents, suggesting a role of Neogenin in forming excitatory synapses. Finally, ablating Neogenin in the BLA in adult male mice impaired fear memory likely by reducing mEPSC frequency in BLA excitatory neurons. These results reveal an unrecognized function of Neogenin in amygdala for information processing by promoting and maintaining neurotransmission and synaptic plasticity and provide insight into molecular mechanisms of neuronal activation in amygdala.SIGNIFICANCE STATEMENT Appropriate neuronal activation in amygdala is critical for information processing. However, the underlying regulatory mechanisms are not well understood. Neogenin is known to regulate axon navigation and adult neurogenesis. Here we show that it is critical for neurotransmission and synaptic plasticity in the amygdala and thus fear memory by using a combination of genetic, electrophysiological, behavioral techniques. Our studies identify a novel function of Neogenin and provide insight into molecular mechanisms of neuronal activation in amygdala for fear processing.


Asunto(s)
Complejo Nuclear Basolateral/metabolismo , Miedo/fisiología , Aprendizaje/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Animales , Potenciales Postsinápticos Excitadores/fisiología , Miedo/psicología , Masculino , Ratones , Ratones Transgénicos , Técnicas de Cultivo de Órganos
7.
J Neuroinflammation ; 16(1): 62, 2019 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-30871577

RESUMEN

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.


Asunto(s)
Antiinflamatorios/uso terapéutico , Cumarinas/uso terapéutico , Citocinas/metabolismo , Encefalitis/tratamiento farmacológico , Regulación de la Expresión Génica/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Citocinas/genética , Modelos Animales de Enfermedad , Encefalitis/etiología , Femenino , Regulación de la Expresión Génica/genética , Gliosis/tratamiento farmacológico , Gliosis/genética , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Placa Amiloide/tratamiento farmacológico , Placa Amiloide/etiología , Presenilina-1/genética , Presenilina-1/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
8.
Brain Behav Immun ; 79: 159-173, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30763768

RESUMEN

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.


Asunto(s)
Janus Quinasa 2/metabolismo , Neurogénesis/fisiología , Factor de Transcripción STAT3/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Desmetilación del ADN , Metilación de ADN , Dioxigenasas/genética , Dioxigenasas/metabolismo , Hipocampo/metabolismo , Humanos , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Transgénicos , Células-Madre Neurales/metabolismo , Neurogénesis/inmunología , Neuroinmunomodulación , Transducción de Señal/inmunología
9.
J Neuroinflammation ; 15(1): 268, 2018 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-30227858

RESUMEN

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.


Asunto(s)
Astrocitos/efectos de los fármacos , Encéfalo/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Infarto de la Arteria Cerebral Media/fisiopatología , Netrina-1/farmacología , Animales , Células Cultivadas , Infarto de la Arteria Cerebral Media/inducido químicamente , Interleucinas/metabolismo , Lipopolisacáridos , Ratones , Netrina-1/metabolismo
10.
Brain Behav Immun ; 69: 351-363, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29253574

RESUMEN

Alzheimer'sdisease(AD) is characterized by deposition of amyloid-ß (Aß)plaques, neurofibrillary tangles, andneuronal loss, accompaniedbyneuroinflammation. Neuroinflammatoryprocesses are thought to contribute toAD pathophysiology. Metformin has been reported to have anti-inflammatory efficacy. However, whether metformin is responsible for the anti-neuroinflammationand neuroprotection on APPswe/PS1ΔE9 (APP/PS1) mice remains unclear. Here we showed that metformin attenuated spatial memory deficit, neuron loss in the hippocampus and enhanced neurogenesis in APP/PS1 mice. In addition, metformin administration decreased amyloid-ß (Aß)plaque load and chronic inflammation (activated microglia and astrocytes as well as pro-inflammatory mediators) in the hippocampus and cortex. Further study demonstrated that treatment with metformin enhanced cerebral AMPK activation. Meanwhile, metformin notably suppressed the activation of P65 NF-κB, mTOR and S6K, reduced Bace1 protein expression. Our data suggest that metformin can exert functional recovery of memory deficits and neuroprotective effect on APP/PS1 mice via triggering neurogenesis and anti-inflammation mediated by regulating AMPK/mTOR/S6K/Bace1 and AMPK/P65 NF-κB signaling pathways in the hippocampus, which may contribute to improvement in neurological deficits.


Asunto(s)
Enfermedad de Alzheimer/patología , Hipocampo/efectos de los fármacos , Trastornos de la Memoria/prevención & control , Metformina/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Placa Amiloide/prevención & control , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Hipocampo/metabolismo , Hipocampo/patología , Masculino , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Metformina/farmacología , Ratones , Ratones Transgénicos , Fármacos Neuroprotectores/farmacología , Placa Amiloide/metabolismo , Placa Amiloide/patología , Presenilina-1/genética , Presenilina-1/metabolismo
11.
Med Sci Monit ; 21: 3608-15, 2015 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-26590375

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is the most common type of dementia. It causes progressive brain disorder involving loss of normal memory and thinking skills. The transplantation of neural stem cells (NSCs) has been reported to improve learning and memory function of AD rats, and protects basal forebrain cholinergic neurons. Nerve growth factor - poly (ethylene glycol) - poly (lactic-co-glycolic acid)-nanoparticles (NGF-PEG-PLGA-NPs) can facilitate the differentiation of NSCs in vitro. This study thus investigated the treatment efficacy of NGF-PEG-PLGA-NPs combining NSC transplantation in AD model rats. MATERIAL AND METHODS: AD rats were prepared by injection of 192IgG-saporin into their lateral ventricles. Embryonic rat NSCs were separated, induced by NGF-PEG-PLGA-NPs in vitro, and were transplanted. The Morris water-maze test was used to evaluate learning and memory function, followed by immunohistochemical staining for basal forebrain cholinergic neurons, hippocampal synaptophysin, and acetylcholine esterase (AchE) fibers. RESULTS: Rats in the combined treatment group had significantly improved spatial learning ability compared to AD model animals (p<0.05). The number of basal forebrain cholinergic neurons, hippocampal synaptophysin, and AchE-positive fibers were all significantly larger than in the NSC-transplantation group, with no difference from control animals. CONCLUSIONS: NGF-PEG-PLGA-NPs plus NSC transplantation can significantly improve learning and memory functions of AD rats, replenish basal forebrain cholinergic neurons, and help form hippocampal synapses and AchE-positive fibers. These findings may offer practical support for and insight into treatment of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/terapia , Nanopartículas/administración & dosificación , Factor de Crecimiento Nervioso/farmacología , Animales , Prosencéfalo Basal/fisiopatología , Encéfalo/fisiopatología , Neuronas Colinérgicas/patología , Modelos Animales de Enfermedad , Femenino , Hipocampo/metabolismo , Aprendizaje , Masculino , Memoria , Nanopartículas/uso terapéutico , Células-Madre Neurales/trasplante , Poliésteres , Polietilenglicoles , Ratas , Ratas Sprague-Dawley
12.
Cell Biosci ; 14(1): 92, 2024 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-39004750

RESUMEN

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.

13.
Carcinogenesis ; 34(2): 446-53, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23125218

RESUMEN

Nickel (Ni) compounds are well-recognized human carcinogens, yet the molecular mechanisms by which they cause human cancer are still not well understood. MicroRNAs (miRNAs), which are small non-coding RNAs, are involved in diverse biological functions and carcinogenesis. In previous study, we identified upregulation of DNA methyltransferase 1 (DNMT1) expression in nickel sulfide (NiS)-transformed human bronchial epithelial (16HBE) cells. Here, we investigated whether some miRNAs are aberrantly expressed and targets DNMT1 in NiS-transformed cells. Our results showed that the expression of miRNA-152 (miR-152) was specifically downregulated in NiS-transformed cells via promoter DNA hypermethylation, whereas ectopic expression of miR-152 in NiS-transformed cells resulted in a marked reduction of DNMT1 expression. Further experiments revealed that miR-152 directly downregulated DNMT1 expression by targeting the 3' untranslated regions of its transcript. Interestingly, treatment of DNMT inhibitor, 5-aza-2-deoxycytidine, or depletion of DNMT1 led to increased miR-152 expression by reversion of promoter hypermethylation, DNMT1 and MeCP2 binding to miR-152 promoter in NiS-transformed cells. Moreover, inhibition of miR-152 expression in 16HBE cells could increase DNMT1 expression and result in an increase in DNA methylation, DNMT1 and MeCP2 binding to miR-152 promoter, indicating an interaction between miR-152 and DNMT1 is regulated by a double-negative circuit. Furthermore, ectopic expression of miR-152 in NiS-transformed cells led to a significant decrease of cell growth. Conversely, inhibition of miR-152 expression in 16HBE cells significantly increased cell growth. Taken together, these observations demonstrate a crucial functional crosstalk between miR-152 and the DNMT1 via a feedback loop involved in NiS-induced malignant transformation.


Asunto(s)
Carcinógenos/farmacología , Transformación Celular Neoplásica/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Metilación de ADN , Retroalimentación Fisiológica/efectos de los fármacos , MicroARNs/genética , Níquel/efectos adversos , Regiones no Traducidas 3'/genética , Apoptosis/efectos de los fármacos , Western Blotting , Bronquios/citología , Bronquios/efectos de los fármacos , Bronquios/metabolismo , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/genética , Células Cultivadas , Inmunoprecipitación de Cromatina , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Humanos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Cell Biosci ; 13(1): 183, 2023 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-37779199

RESUMEN

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.

15.
J Neuroinflammation ; 9: 202, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22898621

RESUMEN

BACKGROUND: Endogenously produced hydrogen sulfide (H(2)S) may have multiple functions in brain. An increasing number of studies have demonstrated its anti-inflammatory effects. In the present study, we investigated the effect of sodium hydrosulfide (NaHS, a H(2)S donor) on cognitive impairment and neuroinflammatory changes induced by injections of Amyloid-ß(1-40) (Aß(1-40)), and explored possible mechanisms of action. METHODS: We injected Aß(1-40) into the hippocampus of rats to mimic rat model of Alzheimer's disease (AD). 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. The expression of interleukin (IL)-1ß and tumor necrosis factor (TNF)-α was measured by enzyme-linked immunosorbent assay (ELISA) and quantitative real-time polymerase chain reaction (qRT-PCR). The expression of Aß(1-40), phospho-p38 mitogen-activated protein kinase (MAPK), phospho-p65 Nuclear factor (NF)-κB, and phospho-c-Jun N-terminal Kinase (JNK) was analyzed by western blot. RESULTS: We demonstrated that pretreatment with NaHS ameliorated learning and memory deficits in an Aß(1-40) rat model of AD. NaHS treatment suppressed Aß(1-40)-induced apoptosis in the CA1 subfield of the hippocampus. Moreover, the over-expression in IL-1ß and TNF-α as well as the extensive astrogliosis and microgliosis in the hippocampus induced by Aß(1-40) were significantly reduced following administration of NaHS. Concomitantly, treatment with NaHS alleviated the levels of p38 MAPK and p65 NF-κB phosphorylation but not JNK phosphorylation that occurred in the Aß(1-40)-injected hippocampus. CONCLUSIONS: These results indicate that NaHS could significantly ameliorate Aß(1-40)-induced spatial learning and memory impairment, apoptosis, and neuroinflammation at least in part via the inhibition of p38 MAPK and p65 NF-κB activity, suggesting that administration of NaHS could provide a therapeutic approach for AD.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/toxicidad , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Sulfuro de Hidrógeno/uso terapéutico , Trastornos de la Memoria/prevención & control , Fragmentos de Péptidos/toxicidad , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/patología , Animales , Hipocampo/patología , Sulfuro de Hidrógeno/farmacología , Inflamación/inducido químicamente , Inflamación/patología , Inflamación/prevención & control , Masculino , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/patología , Distribución Aleatoria , Ratas , Ratas Wistar , Conducta Espacial/efectos de los fármacos
16.
Front Neurosci ; 16: 1060111, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36605552

RESUMEN

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.

17.
Hum Vaccin Immunother ; 18(6): 2121568, 2022 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-36113067

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer , Vacuna BCG , Dendritas , Animales , Ratones , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/terapia , Espinas Dendríticas/inmunología , Espinas Dendríticas/metabolismo , Espinas Dendríticas/patología , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Interleucina-4/metabolismo , Ratones Transgénicos , Vacuna BCG/uso terapéutico , Dendritas/inmunología , Dendritas/metabolismo , Dendritas/patología , Factor de Necrosis Tumoral alfa/metabolismo
18.
Cell Biosci ; 11(1): 29, 2021 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-33536056

RESUMEN

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.

19.
J Comp Neurol ; 528(3): 389-406, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31423581

RESUMEN

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.


Asunto(s)
Red Nerviosa/química , Red Nerviosa/fisiología , Corteza Visual/química , Corteza Visual/fisiología , Vías Visuales/química , Vías Visuales/fisiología , Animales , Hibridación in Situ/métodos , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Campos Visuales/fisiología
20.
Mol Med Rep ; 21(3): 1172-1180, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31922229

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer , Neuronas Colinérgicas , Aprendizaje , Memoria , Células-Madre Neurales , Presenilina-1 , Trasplante de Células Madre , Sinapsis , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/terapia , Amiloide/genética , Amiloide/metabolismo , Animales , Neuronas Colinérgicas/metabolismo , Neuronas Colinérgicas/patología , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Células-Madre Neurales/trasplante , Presenilina-1/biosíntesis , Presenilina-1/genética , Sinapsis/genética , Sinapsis/metabolismo , Sinapsis/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA