Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Hippocampus ; 31(9): 935-956, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33960056

RESUMEN

Neuron-restrictive silencing factor (NRSF) is a zinc-finger transcription factor that regulates expression of a diverse set of genes. However, NRSF function in brain development still remains elusive. In the present study, we generated NRSF-conditional knockout (NRSF-cKO) mice by hGFAP-Cre/loxp system to study the effect of NRSF deficiency on brain development. Results showed that NRSF conditional knockout caused a smaller hippocampus and a thinner granule cell layer (GCL) in mice. Moreover, the reduction and disarrangement of GFAP+ cells in subgranular zone (SGZ) of NRSF-cKO mice was accompanied with the decreased number of premature neurons, neural stem cells (NSCs) and neural progenitor cells (NPCs), as well as compromising the majority of mitotically active cells. The analysis of postnatal development of hippocampus indicated the existence of an abnormality at postnatal day (P) 8, rather than at P1, in NRSF-cKO mice, although the densities of Ki67+ cells with mitotic ability in dentate gyrus were relatively unaffected at P1 and P8. Meanwhile, NRSF deficiency led to abnormal organization of SGZ at P8 during postnatal development. RNA-Seq analysis revealed 79 deregulated genes in hippocampus of NRSF-cKO mice at P8, which were involved in p53 signal transduction, neuron migration and negative regulation of cell proliferation, etc. The deregulation of p53 pathway in NRSF-cKO mice at P1 and P8 was evidenced, of which p21/Cdkn1a was accumulated in a portion of NSCs and NPCs in hippocampus during postnatal development. Together, these results, for the first time, revealed that NRSF could significantly influence the postnatal development of hippocampus, especially the formation of SGZ.


Asunto(s)
Células-Madre Neurales , Neuronas , Animales , Giro Dentado , Hipocampo , Ratones , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Neuronas/fisiología
2.
Cell Mol Neurobiol ; 39(5): 637-650, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30852720

RESUMEN

Reactive microglia clustering around amyloid plaques in brain is a histopathological feature of Alzheimer's disease (AD) and reflects the contribution of neuroinflammation in AD pathogenesis. ß-Amyloid peptide (Aß) has been shown to induce a range of microglial responses including chemotaxis, cytotoxicity and inflammation, but the underlying mechanism is poorly understood. Considering the fundamental role of RhoA/ROCK signaling in cell migration and its broad implication in AD and neuroinflammation, we hypothesized that RhoA/ROCK signaling might be involved in Aß-induced microglial responses. From in vivo mouse models including APP/PS1 transgene and fibrillar Aß stereotactic injection, we observed the elevated expression level of RhoA in reactive microglia. Through a series in vitro cell migration, cytotoxicity and biochemistry assays, we found that RhoA/ROCK signaling plays an essential role in Aß-induced responses of microglial BV2 cells. Small molecular agents Fasudil and Y27632 showed prominent beneficial effects, which implies the therapeutic potential of RhoA/ROCK signaling inhibitors in AD treatment.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Apoptosis/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Inflamación/patología , Microglía/patología , Transducción de Señal , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Antígenos CD/metabolismo , Línea Celular , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Modelos Biológicos , Fármacos Neuroprotectores/farmacología , Agregado de Proteínas/efectos de los fármacos
3.
Exp Cell Res ; 370(1): 103-115, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29908160

RESUMEN

Tau pathology in Alzheimer's disease (AD) includes hyperphosphorylation and truncation of tau. Phosphorylation at S422 is found to suppress truncation of tau at D421 that leading to the generation of ΔTau. However, the interrelation between hyperphosphorylation and generation of ΔTau in AD remains elusive. In current study, staurosporine (Stau) induced ΔTau generation by caspases in SH-SY5Y cells with tau overexpression was found to be accompanied by a dramatic dephosphorylation at S422 and the epitope of the diagnostic antibody AT8 (S199 + S202 + T205), but a moderate dephosphorylation of PHF1 (S396 + S404) epitope. Therefore, to explore the effect of AT8 epitope on tau truncation, the residues in AT8 epitope were mutated to produce "pseudo-phosphorylated" (AT8E) or "pseudo-unphosphorylated" (AT8A) tau constructs. With Stau treatment, the generation of ΔTau from tau-AT8E was significantly attenuated comparing with that from tau-AT8A, which was S422-independent in that addition of S422A mutation still preserved this effect. Interestingly, this modulatory effect was able to be reversed by addition of PHF1E mutation. Moreover, treating the crude tau extracts with recombinant caspase-3 in vitro, also showed that ΔTau level was suppressed by AT8E, and potentiated by AT8E + PHF1E. The results primarily revealed the modulating effects of phosphorylation on ΔTau generation which may have potential implications in tau pathological processes and therapeutic intervention.


Asunto(s)
Ácido Aspártico/metabolismo , Epítopos/metabolismo , Fosforilación/fisiología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Caspasas/metabolismo , Línea Celular Tumoral , Humanos , Neuronas/metabolismo
4.
Neural Plast ; 2019: 4168472, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30906318

RESUMEN

Exposure to chronic psychiatric stress has been linked to Alzheimer's disease-related tau hyperphosphorylation and abnormalities in glutamate neurotransmission. However, the pathological relationship between glutamatergic dysfunction and tau phosphorylation in the cerebral cortex under chronic psychiatric stress is not fully understood. The present study investigated the effects of memantine (MEM, 5 and 10 mg/kg), an uncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist, on chronic restraint stress- (CRS-) induced tau phosphorylation in mice. CRS administered for 16 or 28 consecutive days (1 h daily) induced significant tau phosphorylation in the brain. MEM treatment suppressed the elevation of phosphorylated tau (P-tau) levels induced by 16-day CRS in a dose-dependent manner. P-tau reduction was accompanied by the attenuation of the upregulation of GSK3ß and CDK5 expression and the downregulation of PP2A activity induced by CRS. Additionally, MEM reduced CRS-induced upregulation of NMDA receptor subunit levels (GluN2A, GluN2B) in the frontal cortex. However, MEM markedly enhanced tau phosphorylation in the frontal cortex and other cerebral cortical regions following 28 days of CRS. The stimulatory effect of MEM on CRS-induced tau phosphorylation was correlated with increased activities of AKT, JNK, and GSK3ß, inactivation of PP2A, and downregulation of Pin1 and HSP70. Moreover, MEM did not effectively reverse the NMDA receptor upregulation induced by 28-day CRS and even increased GluN2B subunit levels. In contrast to the duration-dependent effects of MEM on P-tau levels, MEM produced an anxiolytic effect in both regimens as revealed by elevated plus maze testing. However, MEM did not affect the body weight reduction induced by CRS. Thus, MEM exerts distinctive effects on CRS-induced tau phosphorylation, which might be related to the expression of GluN2B. The differential effects of MEM on P-tau levels have crucial implications for its clinical application.


Asunto(s)
Encéfalo/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Memantina/farmacología , Neuronas/efectos de los fármacos , Estrés Psicológico/metabolismo , Proteínas tau/metabolismo , Animales , Encéfalo/metabolismo , Ratones , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Restricción Física , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
5.
Acta Pharmacol Sin ; 39(10): 1582-1589, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29795362

RESUMEN

Both in vivo and in vitro studies have shown the beneficial effects of the delta-opioid receptor (DOR) on neurodegeneration in hypoxia/ischemia. We previously reported that DOR stimulation with [(D-Ala2, D-Leu5) enkephalin] (DADLE), a potent DOR agonist, for both a short (minutes) and long (days) time has notable protective effects against sodium azide (NaN3)-induced cell injury in primary cultured rat cortical neurons. We further demonstrated that short-term DADLE stimulation increased neuronal survival through the PKC-mitochondrial ERK pathway. However, the mechanisms underlying long-term neuroprotection by DADLE remain unclear. Here, we showed that DOR stimulation with DADLE (0.1 µmol/L) for 2 d selectively activates the PI3K/Akt/NF-κB pathway in NaN3-treated neurons; this activation increased Bcl-2 expression, attenuated Cyto c release and promoted neuronal survival. Further investigation revealed that sustained DADLE stimulation increased Bcl-2 expression by enhancing NF-κB binding to the Bcl-2 promoter and upregulating the histone acetylation levels of the Bcl-2 promoter. Our results demonstrate that prolonged DADLE exposure epigenetically promotes Bcl-2 expression and elicits neuroprotective effects in the NaN3 model via the PI3K/Akt/NF-κB pathway.


Asunto(s)
Leucina Encefalina-2-Alanina/farmacología , Epigénesis Genética/efectos de los fármacos , Neuroprotección/fisiología , Fármacos Neuroprotectores/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Transducción de Señal/efectos de los fármacos , Animales , Células Cultivadas , Citocromos c/metabolismo , FN-kappa B/metabolismo , Neuronas/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Sprague-Dawley , Receptores Opioides delta/agonistas , Regulación hacia Arriba
6.
Sheng Li Xue Bao ; 66(2): 107-17, 2014 Apr 25.
Artículo en Zh | MEDLINE | ID: mdl-24777400

RESUMEN

To investigate the murine double minute 2 (MDM2) localization and expression pattern in brain, immunohistochemistry, immunofluorescent staining and immunoblotting methods were used to analyze it in brains of Kunming mice during postnatal development, in brains of adult SD rats and in primarily cultured neurons. The distribution of MDM2 and markers of axon initial segment (AIS) was analyzed by double immunolabeling. In addition, Nutlin-3, a MDM2 antagonist, was injected into hippocampus to analyze the effect on the distribution of MDM2 and AIS protein Nav1.6 in AIS. The results showed that the dynamic expression patterns of MDM2 protein in cerebral cortex and hippocampus of Kunming mice after birth were different. However, it was similar that MDM2 was gradually enriched to AIS during postnatal development, especially after postnatal day 7. The MDM2 in AIS was also observed in different brain regions of adult SD rat brain and in primarily cultured neurons, where MDM2 was colocalized with AIS markers such as AnkG and Nav1.6. In addition, hippocampal injection of Nutlin-3 could induce the loss of the characteristic distribution of MDM2 in AIS. Moreover, Nutlin-3 not only caused a decrease of Nav1.6 distributing in AIS, but also disrupted the polarized distribution of MAP2 in neurons. These results indicate that MDM2 can be enriched at the AIS of adult rodent brain, which might play a role in regulation of the maintenance of AIS function and neuronal polarity.


Asunto(s)
Axones/metabolismo , Corteza Cerebral/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Animales , Imidazoles/farmacología , Ratones , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Piperazinas/farmacología , Ratas , Ratas Sprague-Dawley
7.
Brain Sci ; 14(5)2024 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-38790466

RESUMEN

BACKGROUND: The discovery of novel diagnostic methods and therapies for Alzheimer's disease (AD) faces significant challenges. Previous research has shed light on the neuroprotective properties of Apelin-13 in neurodegenerative disorders. However, elucidating the mechanism underlying its efficacy in combating AD-related nerve injury is imperative. In this study, we aimed to investigate Apelin-13's mechanism of action in an in vivo model of AD induced by streptozocin (STZ). METHODS: We utilized an STZ-induced nerve injury model of AD in mice to investigate the effects of Apelin-13 administration. Apelin-13 was administered intranasally, and cognitive impairment was assessed using standardized behavioral tests, primarily, behavioral assessment, histological analysis, and biochemical assays, in order to evaluate synaptic plasticity and oxidative stress signaling pathways. RESULTS: Our findings indicate that intranasal administration of Apelin-13 ameliorated cognitive impairment in the STZ-induced AD model. Furthermore, we observed that this effect was potentially mediated by the enhancement of synaptic plasticity and the attenuation of oxidative stress signaling pathways. CONCLUSIONS: The results of this study suggest that intranasal administration of Apelin-13 holds promise as a therapeutic strategy for preventing neurodegenerative diseases such as AD. By improving synaptic plasticity and mitigating oxidative stress, Apelin-13 may offer a novel approach to neuroprotection in AD and related conditions.

8.
Neuroscience ; 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38960088

RESUMEN

Hippocampus is a critical component of the central nervous system. SRSF10 is expressed in central nervous system and plays important roles in maintaining normal brain functions. However, its role in hippocampus development is unknown. In this study, using SRSF10 conditional knock-out mice in neural progenitor cells (NPCs), we found that dysfunction of SRSF10 leads to developmental defects in the dentate gyrus of hippocampus, which manifests as the reduced length and wider suprapyramidal blade and infrapyramidal blade.Furthermore, we proved that loss of SRSF10 in NPCs caused inhibition of the differentiation activity and the abnormal migration of NPCs and granule cells, resulting in reduced granule cells and more ectopic granule cells dispersed in the molecular layer and hilus. Finally, we found that the abnormal migration may be caused by the radial glia scaffold and the reduced DISC1 expression in NPCs. Together, our results indicate that SRSF10 is required for the cell migration and formation of dentate gyrus during the development of hippocampus.

9.
J Biol Inorg Chem ; 18(1): 39-47, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23086305

RESUMEN

Aggregation and cytotoxicity of Aß with redox-active metals in neuronal cells have been implicated in the progression of Alzheimer disease. Human metallothionein (MT) 3 is highly expressed in the normal human brain and is downregulated in Alzheimer disease. Zn(7)MT3 can protect against the neuronal toxicity of Aß by preventing copper-mediated Aß aggregation, abolishing the production of reactive oxygen species (ROS) and the related cellular toxicity. In this study, we intended to decipher the roles of single-domain proteins (α/ß) and the α-ß domain-domain interaction of Zn(7)MT3 to determine the molecular mechanism for protection against the neuronal cytotoxicity of Aß(1-42) with copper ions. With this in mind, the α and ß single-domain proteins, heterozygous ß(MT3)-α(MT1), and a linker-truncated mutant ∆31-34 were prepared and characterized. In the presence/absence of various Zn(7)MT3 proteins, the Aß(1-42)-Cu(2+)-mediated aggregation, the production of ROS, and the cellular toxicity were investigated by transmission electron microscopy, ROS assay by means of a fluorescent probe, and SH-SY5Y cell viability, respectively. The ß domain cannot abolish Aß(1-42)-Cu(2+)-induced aggregation, and neither the ß domain nor the α domain can quench the production of ROS because of the redox cycling of Aß-Cu(2+). Similarly to wild-type Zn(7)MT3, the heterozygous ß(MT3)-α(MT1) possesses the characteristic of alleviating Aß(1-42) aggregation and oxidative stress to neuronal cells. Therefore, the two domains through the linker Lys-Lys-Ser form a cooperative unit, and each of them is indispensable in conducting its bioactivity. The α domain plays an important role in modulating the stability of the metal-thiolate cluster, and the α-ß domain-domain interaction through the linker is critical for its protective role in the brain.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Cobre/toxicidad , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fragmentos de Péptidos/metabolismo , Péptidos beta-Amiloides/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Metalotioneína 3 , Proteínas del Tejido Nervioso/química , Neuronas/citología , Fragmentos de Péptidos/química , Multimerización de Proteína/efectos de los fármacos , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Especies Reactivas de Oxígeno/metabolismo
10.
Int J Neurosci ; 123(11): 783-91, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23668913

RESUMEN

Estrogen has beneficial effects on neurodegenerative disorders and cognitive function of postmenopausal women. Puerarin, isolated from Pueraria lobota, has been classified as a phytoestrogen, which can be highly effective against cerebrovascular diseases. In this study, the effects of puerarin on neural cholinergic system in the brain of ovariectomized guinea pigs were studied. The puerarin at the doses used (15 mg/kg body weight (bw)/day and 30 mg/kg bw/day) for 10 days had the estrogenic activity indicated by the attenuation of the reduction of uterine weight induced by ovariectomy. In brain, puerarin treatment increased choline acetyltransferase (ChAT) activity and expression in hippocampus, and increased ChAT immnuopositive signals in septal diagonal region. Puerarin treatment could suppress the increase of acetylcholinesterase expression and activity to the levels of the intact group, although they were not significantly different from those of the ovariectomized animals. Moreover, puerarin decreased the ß-amyloid immunopositive staining in hippocampus. In brief, the present study suggests that puerarin prevents the dysfunction of the neuronal cholinergic system and ameliorates the increase of ß-amyloid caused by estrogen deficiency.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Colina O-Acetiltransferasa/metabolismo , Isoflavonas/farmacología , Ovariectomía , Animales , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Femenino , Cobayas , Ovariectomía/tendencias
11.
Sheng Li Xue Bao ; 65(3): 253-62, 2013 Jun 25.
Artículo en Zh | MEDLINE | ID: mdl-23788181

RESUMEN

Small ubiquitin-related modifiers (SUMOs) belong to an important class of ubiquitin like proteins. SUMOylation is a post-translational modification process that regulates the functional properties of many proteins, among which are several proteins implicated in neurodegenerative diseases. This study was aimed to investigate the changes of SUMO-1 expression and modification, and the relationship between SUMO-1 and Alzheimer's disease (AD) pathology in APP/PS1 transgenic AD mice. Using Western blot, co-immunoprecipitation and immunofluorescent staining methods, the SUMO-1 expression and modification and its relation to tau, amyloid precursor protein (APP) and ß-amyloid protein (Aß) in the 12-month-old APP/PS1 transgenic AD mice were analyzed. The results showed that: (1) Compared with the normal wild-type mice, the expression and modification of SUMO-1 increased in brain of AD mice, which was accompanied by an increase of ubiquitination; (2) In RIPA soluble protein fraction of cerebral cortex, co-immunoprecipitation analysis showed tau SUMOylated by SUMO-1 increased in AD mice, however, AT8 antibody labeled phosphorylated tau was less SUMOylated whereas PS422 antibody labeled phosphorylated tau was similar to control mice; (3) Double immunofluorescent staining showed that SUMO-1 could distributed in amyloid plaques, appearing that some of SUMO-1 diffused in centre of some plaques and some of SUMO-1 co-localized with AT8 labeled phosphorylated tau forming punctate aggregates around amyloid plaques which was concerned as dystrophic neurites, however, less Aß, APP and PS422 labeled phosphorylated tau were found co-localized with SUMO-1. These results suggest that SUMO-1 expression and modification increase abnormally in transgenic AD mice, which may participate in modulation of the formation of senile plaques and dystrophic neurites.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Placa Amiloide/fisiopatología , Proteína SUMO-1/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/patología , Ratones , Ratones Transgénicos , Neuritas/patología , Fosforilación , Sumoilación , Proteínas tau/metabolismo
12.
iScience ; 26(7): 107042, 2023 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-37360696

RESUMEN

Alternative pre-mRNA splicing plays critical roles in brain development. SRSF10 is a splicing factor highly expressed in central nervous system and plays important roles in maintaining normal brain functions. However, its role in neural development is unclear. In this study, by conditional depleting SRSF10 in neural progenitor cells (NPCs) in vivo and in vitro, we found that dysfunction of SRSF10 leads to developmental defects of the brain, which manifest as abnormal ventricle enlargement and cortical thinning anatomically, as well as decreased NPCs proliferation and weakened cortical neurogenesis histologically. Furthermore, we proved that the function of SRSF10 on NPCs proliferation involved the regulation of PI3K-AKT-mTOR-CCND2 pathway and the alternative splicing of Nasp, a gene encoding isoforms of cell cycle regulators. These findings highlight the necessity of SRSF10 in the formation of a structurally and functionally normal brain.

13.
Int J Neurosci ; 122(6): 290-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22185089

RESUMEN

Accumulation of the amyloid ß protein (Aß) in the brain is an important step in the pathogenesis of Alzheimer's disease. Many molecules could bind with Aß, among which some molecules mediate Aß neuronal toxicity. Thus, it is of interest to study the binding proteins of Aß, and the functions that might be affected by Aß. In the present study, we observed that accumulation of α-subunit of ATP synthase is associated with aggregates of Aß proteins in amyloid plaques of amyloid precursor protein/presennillin-1 transgenic mice, and identified the α-subunit of ATP synthase as one of the Aß binding proteins on the plasma membrane of neural cells by Western blot and mass spectrometry. In order to evaluate the consequences of the interaction between Aß and surface α-subunit of ATP synthase, the extracellular ATP generation was analyzed, which showed that aggregated Aß partially inhibited the extracellular generation of ATP, but was unable to significantly induce a decrease in cell surface ATP synthase α on neurons. These results suggest that the cell surface ATP synthase α is a binding protein for Aß on neural cells, the functional inhibition of surface ATP synthase might be involved in machinery of brain malfunction in Aß-mediated pathogenesis of Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Adenosina Trifosfato/biosíntesis , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/metabolismo , Encéfalo/ultraestructura , Células Cultivadas , Femenino , Masculino , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Neuronas/ultraestructura , Fragmentos de Péptidos/metabolismo , Placa Amiloide/metabolismo , Presenilina-1/genética , Ratas , Ratas Sprague-Dawley
14.
Front Aging Neurosci ; 14: 1057281, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36589543

RESUMEN

Introduction: Hyperphosphorylated Tau formed neurofibrillary tangles was one of the major neuropathological hallmarks of Alzheimer's disease (AD). Dysfunctional insulin signaling in brain is involved in AD. However, the effect of Tau pathology on brain insulin resistance remains unclear. This study explored the effects of overexpressing wild-type Tau (WTau) or Tau with pseudo-phosphorylation at AT8 residues (PTau) on the insulin signaling pathway (ISP). Methods: 293T cells or SY5Y cells overexpressing WTau or PTau were treated with or without insulin. The elements in ISP or the regulators of IPS were analyzed by immunoblotting, immunofluorescent staining and co-immunoprecipitation. Akt inhibitor MK2206 was used for evaluating the insulin signaling to downstream of mTOR in Tau overexpressing cells. The effects of anti-aging drug lonafarnib on ISP in WTau or PTau cells were also analyzed with immunoblotting. Considering lonafarnib is an inhibitor of FTase, the states of Rhes, one of FTase substrate in WTau or PTau cells were analyzed by drug affinity responsive target stability (DARTS) assay and the cellular thermal shift assay (CETSA). Results: WTau or PTau overexpression in cells upregulated basal activity of elements in ISP in general. However, overexpression of WTau or PTau suppressed the ISP signaling transmission responses induced by insulin simulation, appearing relative higher response of IRS-1 phosphorylation at tyrosine 612 (IRS-1 p612) in upstream IPS, but a lower phosphorylation response of downstream IPS including mTOR, and its targets 4EPB1 and S6. This dysregulation of insulin evoked signaling transmission was more obvious in PTau cells. Suppressing Akt with MK2206 could compromise the levels of p-S6 and p-mTOR in WTau or PTau cells. Moreover, the changes of phosphatases detected in WTau and PTau cells may be related to ISP dysfunction. In addition, the effects of lonafarnib on the ISP in SY5Y cells with WTau and PTau overexpression were tested, which showed that lonafarnib treatment resulted in reducing the active levels of ISP elements in PTau cells but not in WTau cells. The differential effects are probably due to Tau phosphorylation modulating lonafarnib-induced alterations in Rhes, as revealed by DARTS assay. Conclusion and discussion: Overexpression of Tau or Tau with pseudo-phosphorylation at AT8 residues could cause an upregulation of the basal/tonic ISP, but a suppression of insulin induced the phasic activation of ISP. This dysfunction of ISP was more obvious in cells overexpressing pseudo-phosphorylated Tau. These results implied that the dysfunction of ISP caused by Tau overexpression might impair the physiological fluctuation of neuronal functions in AD. The different effects of lonafarnib on ISP between WTau and PTau cells, indicating that Tau phosphorylation mediates an additional effect on ISP. This study provided a potential linkage of abnormal expression and phosphorylation of Tau to the ISP dysfunction in AD.

15.
J Biol Chem ; 285(49): 38214-23, 2010 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-20889974

RESUMEN

Parkin is an E3 ubiquitin ligase that mediates the ubiquitination of protein substrates. The mutations in the parkin gene can lead to a loss of function of parkin and cause autosomal recessive juvenile onset parkinsonism. Recently, parkin was reported to be involved in the regulation of mitophagy. Here, we identify the Bcl-2, an anti-apoptotic and autophagy inhibitory protein, as a substrate for parkin. Parkin directly binds to Bcl-2 via its C terminus and mediates the mono-ubiquitination of Bcl-2, which increases the steady-state levels of Bcl-2. Overexpression of parkin, but not its ligase-deficient forms, decreases autophagy marker LC3 conversion, whereas knockdown of parkin increases LC3 II levels. In HeLa cells, a parkin-deficient cell line, knockdown of parkin does not change LC3 conversion. Moreover, overexpression of parkin enhances the interactions between Bcl-2 and Beclin 1. Our results provide evidence that parkin mono-ubiquitinates Bcl-2 and regulates autophagy via Bcl-2.


Asunto(s)
Autofagia , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Trastornos Distónicos/congénito , Trastornos Distónicos/genética , Trastornos Distónicos/metabolismo , Células HeLa , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-bcl-2/genética , Ubiquitina-Proteína Ligasas/genética
16.
J Biol Chem ; 285(49): 38630-40, 2010 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-20921223

RESUMEN

Dysbindin-1 is a 50-kDa coiled-coil-containing protein encoded by the gene DTNBP1 (dystrobrevin-binding protein 1), a candidate genetic factor for schizophrenia. Genetic variations in this gene confer a susceptibility to schizophrenia through a decreased expression of dysbindin-1. It was reported that dysbindin-1 regulates the expression of presynaptic proteins and the release of neurotransmitters. However, the precise functions of dysbindin-1 are largely unknown. Here, we show that dysbindin-1 is a novel nucleocytoplasmic shuttling protein and translocated to the nucleus upon treatment with leptomycin B, an inhibitor of exportin-1/CRM1-mediated nuclear export. Dysbindin-1 harbors a functional nuclear export signal necessary for its nuclear export, and the nucleocytoplasmic shuttling of dysbindin-1 affects its regulation of synapsin I expression. In brains of sandy mice, a dysbindin-1-null strain that displays abnormal behaviors related to schizophrenia, the protein and mRNA levels of synapsin I are decreased. These findings demonstrate that the nucleocytoplasmic shuttling of dysbindin-1 regulates synapsin I expression and thus may be involved in the pathogenesis of schizophrenia.


Asunto(s)
Proteínas Portadoras/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Regulación de la Expresión Génica , Esquizofrenia/metabolismo , Sinapsinas/biosíntesis , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/genética , Animales , Antibióticos Antineoplásicos/farmacología , Encéfalo/metabolismo , Proteínas Portadoras/genética , Núcleo Celular/genética , Citoplasma/genética , Disbindina , Proteínas Asociadas a la Distrofina , Ácidos Grasos Insaturados/farmacología , Células HEK293 , Humanos , Carioferinas/antagonistas & inhibidores , Carioferinas/genética , Carioferinas/metabolismo , Ratones , Ratones Mutantes , Terminales Presinápticos/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Esquizofrenia/genética , Sinapsinas/genética , Proteína Exportina 1
17.
J Biol Inorg Chem ; 16(5): 809-16, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21523435

RESUMEN

The ß-amyloid peptide (Aß) aggregation in the brain, known as amyloid plaques, is a hallmark of Alzheimer's disease (AD). The aberrant interaction of Cu(2+) ion with Aß potentiates AD by inducing Aß aggregation and generating neurotoxic reactive oxygen species (ROS). In this study, the biosynthesized recombinant Aß(1-40) was, for the first time, used to investigate the mechanism for heme to prevent Aß(1-40) aggregation and its cytotoxicity. Cell viability studies of SH-SY5Y cells and rat primary hippocampal neurons showed that exogenous heme can protect the cells by reducing cytotoxicity in the presence of Cu(2+) and/or Aß(1-40). UV-vis spectroscopy, circular dichroism spectroscopy, and differential pulse voltammetry were applied to examine the interaction between heme and Aß(1-40). It was proven that a heme-Aß(1-40) complex is formed and can stabilize the α-helix structure of Aß(1-40) to inhibit Aß(1-40) aggregation. The heme-Aß(1-40) complex possesses peroxidase activity and it may catalyze the decomposition of H(2)O(2), reduce the generation of ROS downstream, and ultimately protect the cells. These results indicated that exogenous heme is able to alleviate the cytotoxicity induced by Aß(1-40) and Cu(2+). This information may be a foundation to develop a potential strategy to treat AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Neuronas/metabolismo , Fragmentos de Péptidos/metabolismo , Animales , Línea Celular , Supervivencia Celular , Células Cultivadas , Cobre/metabolismo , Hemo/metabolismo , Hipocampo/citología , Humanos , Estrés Oxidativo , Peroxidasa/metabolismo , Ratas , Proteínas Recombinantes/metabolismo
18.
Cell Biol Int ; 35(1): 81-6, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20626349

RESUMEN

The ATP synthase is known to play important roles in ATP generation and proton translocation within mitochondria. Here, we now provide evidence showing the presence of functional ecto-ATP synthase on the neuronal surface. Immunoblotting revealed that the α, ß subunits of ATP synthase F1 portion are present in isolated fractions of plasma membrane and biotin-labelled surface protein from primary cultured neurons; the surface distribution of α, ß subunits was also confirmed by immunofluorescence staining. Moreover, α and ß subunits were also found in fractions of plasma membrane and lipid rafts isolated from rat brain, and flow cytometry analysis showed α subunits on the surface of acutely isolated brain cells. Activity assays showed that the extracellular ATP generation of cultured neurons could be compromised by α, ß subunit antibodies and ATP synthase inhibitors. pH(i) (intracellular pH) analysis demonstrated that at low extracellular pH, α or ß subunit antibodies decreased pHi of primary cultured neurons. Therefore, ATP synthase on the surface of neurons may be involved in the machineries of extracellular ATP generation and pH(i) homoeostasis.


Asunto(s)
Complejos de ATP Sintetasa/fisiología , Equilibrio Ácido-Base , Adenosina Trifosfato/biosíntesis , Líquido Extracelular/metabolismo , Neuronas/metabolismo , Animales , Membrana Celular/enzimología , Células Cultivadas , Embrión de Mamíferos/citología , Femenino , Concentración de Iones de Hidrógeno , Masculino , Neuronas/citología , Ratas , Ratas Sprague-Dawley
19.
Neurosci Lett ; 758: 136005, 2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34098024

RESUMEN

Neuroinflammation is one of the main causes of Alzheimer's disease (AD). The presence of Lipopolysaccharide (LPS) in senile plaques (SP) of AD suggests that it plays a role in AD pathogenesis. ATP5A1 (F1F0-ATP synthase F1 α subunit) is abundant in SP. Further, the protein has recently been found to have an anti-infection role in zebrafish embryos. In the present study, we observed that LPS levels were higher in the brains of APP/PS1 mice than in control mice, and LPS co-localised with ATP5A1 in amyloid plaques. The interaction of recombinant ATP5A1(rATP5A1) and LPS was evidenced by cellular thermal shift assay and enzyme-linked immunosorbent assay-based binding assay in vitro. Neuroinflammation in the brain of a mouse model was induced by intracerebroventricular injection of LPS. The addition of rATP5A1 relieved LPS-induced reduction of spontaneous locomotor ability, depressive-like behaviour, and working memory impairment. Furthermore, rATP5A1 suppressed the activation of astrocytes and microglia, IL-1ß accumulation, and tau phosphorylation induced by LPS. Taken together, findings suggest that ATP5A1 is involved in the regulation of LPS-mediated neuroinflammation in AD.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Encéfalo/patología , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Enfermedades Neuroinflamatorias/inmunología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/inmunología , Modelos Animales de Enfermedad , Humanos , Inyecciones Intraventriculares , Interleucina-1beta/metabolismo , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/inmunología , Lipopolisacáridos/metabolismo , Masculino , Ratones , Ratones Transgénicos , ATPasas de Translocación de Protón Mitocondriales/administración & dosificación , Mutación , Enfermedades Neuroinflamatorias/genética , Enfermedades Neuroinflamatorias/patología , Fosforilación/efectos de los fármacos , Presenilina-1/genética , Unión Proteica , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/metabolismo
20.
Nat Metab ; 3(11): 1536-1551, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34782792

RESUMEN

Beiging of white adipose tissue (WAT) is associated with an increase of anti-inflammatory M2-like macrophages in WAT. However, mechanisms through which M2-like macrophages affect beiging are incompletely understood. Here, we show that the macrophage cytokine Slit3 is secreted by adipose tissue macrophages and promotes cold adaptation by stimulating sympathetic innervation and thermogenesis in mice. Analysing the transcriptome of M2-like macrophages in murine inguinal WAT (iWAT) after cold exposure, we identify Slit3 as a secreted cytokine. Slit3 binds to the ROBO1 receptor on sympathetic neurons to stimulate Ca2+/calmodulin-dependent protein kinase II signalling and norepinephrine release, which enhances adipocyte thermogenesis. Adoptive transfer of Slit3-overexpressing M2 macrophages to iWAT promotes beiging and thermogenesis, whereas mice that lack Slit3 in myeloid cells are cold-intolerant and gain more weight. Our findings shed new light on the integral role of M2-like macrophages for adipose tissue homeostasis and uncover the macrophage-Slit3-sympathetic neuron-adipocyte signalling axis as a regulator of long-term cold adaptation.


Asunto(s)
Tejido Adiposo/inervación , Tejido Adiposo/fisiología , Fibras Adrenérgicas/fisiología , Macrófagos/metabolismo , Proteínas de la Membrana/biosíntesis , Termogénesis , Tejido Adiposo Blanco/inervación , Tejido Adiposo Blanco/metabolismo , Animales , Plasticidad de la Célula , Metabolismo Energético , Regulación de la Expresión Génica , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Especificidad de Órganos/genética , Fosforilación , Unión Proteica , Receptores Inmunológicos/metabolismo , Temperatura , Termogénesis/genética , Proteínas Roundabout
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA