Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neural Regen Res ; 18(3): 587-593, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36018182

RESUMEN

Mild cognitive impairment (MCI) is a prodrome of Alzheimer's disease pathology. Cognitive impairment patients often have a delayed diagnosis because there are no early symptoms or conventional diagnostic methods. Exosomes play a vital role in cell-to-cell communications and can act as promising biomarkers in diagnosing diseases. This study was designed to identify serum exosomal candidate proteins that may play roles in diagnosing MCI. Mass spectrometry coupled with tandem mass tag approach-based non-targeted proteomics was used to show the differentially expressed proteins in exosomes between MCI patients and healthy controls, and these differential proteins were validated using immunoblot and enzyme-linked immunosorbent assays. Correlation of cognitive performance with the serum exosomal protein level was determined. Nanoparticle tracking analysis suggested that there was a higher serum exosome concentration and smaller exosome diameter in individuals with MCI compared with healthy controls. We identified 69 exosomal proteins that were differentially expressed between MCI patients and healthy controls using mass spectrometry analysis. Thirty-nine exosomal proteins were upregulated in MCI patients compared with those in control patients. Exosomal fibulin-1, with an area under the curve value of 0.81, may be a biomarker for an MCI diagnosis. The exosomal protein signature from MCI patients reflected the cell adhesion molecule category. In particular, higher exosomal fibulin-1 levels correlated with lower cognitive performance. Thus, this study revealed that exosomal fibulin-1 is a promising biomarker for diagnosing MCI.

2.
Mol Med ; 28(1): 11, 2022 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-35093024

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is the most common type of neurodegenerative disease in the contemporary era, and it is still clinically incurable. Eriodictyol, a natural flavonoid compound that is mainly present in citrus fruits and some Chinese herbal medicines, has been reported to exert anti-inflammatory, antioxidant, anticancer and neuroprotective effects. However, few studies have examined the anti-AD effect and molecular mechanism of eriodictyol. METHODS: APP/PS1 mice were treated with eriodictyol and the cognitive function of mice was assessed using behavioral tests. The level of amyloid-ß (Aß) aggregation and hyperphosphorylation of Tau in the mouse brain were detected by preforming a histological analysis and Western blotting. HT-22 cells induced by amyloid-ß peptide (1-42) (Aß1-42) oligomers were treated with eriodictyol, after which cell viability was determined and the production of p-Tau was tested using Western blotting. Then, the characteristics of ferroptosis, including iron aggregation, lipid peroxidation and the expression of glutathione peroxidase type 4 (GPX4), were determined both in vivo and in vitro using Fe straining, Western blotting and qPCR assays. Additionally, the expression level of vitamin D receptor (VDR) and the nuclear factor erythroid 2-related factor 2/heme oxygenase-1 (Nrf2/HO-1) signaling pathway were tested using Western blotting and qPCR assays. Afterward, HT-22 cells with VDR knockout were used to explore the potential mechanisms, and the relationship between VDR and Nrf2 was further assessed by performing a coimmunoprecipitation assay and bioinformatics analysis. RESULTS: Eriodictyol obviously ameliorated cognitive deficits in APP/PS1 mice, and suppressed Aß aggregation and Tau phosphorylation in the brains of APP/PS1 mice. Moreover, eriodictyol inhibited Tau hyperphosphorylation and neurotoxicity in HT-22 cells induced by Aß1-42 oligomer. Furthermore, eriodictyol exerted an antiferroptosis effect both in vivo and in vitro, and its mechanism may be associated with the activation of the Nrf2/HO-1 signaling pathway. Additionally, further experiments explained that the activation of Nrf2/HO-1 signaling pathway by eriodictyol treatment mediated by VDR. CONCLUSIONS: Eriodictyol alleviated memory impairment and AD-like pathological changes by activating the Nrf2/HO-1 signaling pathway through a mechanism mediated by VDR, which provides a new possibility for the treatment of AD.


Asunto(s)
Ferroptosis/efectos de los fármacos , Flavanonas/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Receptores de Calcitriol/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Biomarcadores , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Flavanonas/química , Inmunohistoquímica , Ratones , Ratones Transgénicos , Fosforilación , Agregación Patológica de Proteínas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Proteínas tau/metabolismo
3.
Aging Cell ; 18(4): e12961, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31012223

RESUMEN

Cav1.2 is the pore-forming subunit of L-type voltage-gated calcium channel (LTCC) that plays an important role in calcium overload and cell death in Alzheimer's disease. LTCC activity can be regulated by estrogen, a sex steroid hormone that is neuroprotective. Here, we investigated the potential mechanisms in estrogen-mediated regulation of Cav1.2 protein. We found that in cultured primary neurons, 17ß-estradiol (E2) reduced Cav1.2 protein through estrogen receptor α (ERα). This effect was offset by a proteasomal inhibitor MG132, indicating that ubiquitin-proteasome system was involved. Consistently, the ubiquitin (UB) mutant at lysine 29 (K29R) or the K29-deubiquitinating enzyme TRAF-binding protein domain (TRABID) attenuated the effect of ERα on Cav1.2. We further identified that the E3 ligase Mdm2 (double minute 2 protein) and the PEST sequence in Cav1.2 protein played a role, as Mdm2 overexpression and the membrane-permeable PEST peptides prevented ERα-mediated Cav1.2 reduction, and Mdm2 overexpression led to the reduced Cav1.2 protein and the increased colocalization of Cav1.2 with ubiquitin in cortical neurons in vivo. In ovariectomized (OVX) APP/PS1 mice, administration of ERα agonist PPT reduced cerebral Cav1.2 protein, increased Cav1.2 ubiquitination, and improved cognitive performances. Taken together, ERα-induced Cav1.2 degradation involved K29-linked UB chains and the E3 ligase Mdm2, which might play a role in cognitive improvement in OVX APP/PS1 mice.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Canales de Calcio Tipo L/metabolismo , Receptor alfa de Estrógeno/metabolismo , Neuronas/metabolismo , Oligopéptidos/genética , Proteolisis/efectos de los fármacos , Ubiquitinación/efectos de los fármacos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Animales , Línea Celular Tumoral , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Estradiol/farmacología , Receptor alfa de Estrógeno/agonistas , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Leupeptinas/farmacología , Ratones/embriología , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenoles/farmacología , Fenoles/uso terapéutico , Inhibidores de Proteasoma/farmacología , Proteínas Proto-Oncogénicas c-mdm2/genética , Pirazoles/farmacología , Pirazoles/uso terapéutico , Transfección , Ubiquitina/metabolismo
4.
Brain ; 142(1): 176-192, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30596903

RESUMEN

MMP13 (matrix metallopeptidase 13) plays a key role in bone metabolism and cancer development, but has no known functions in Alzheimer's disease. In this study, we used high-throughput small molecule screening in SH-SY5Y cells that stably expressed a luciferase reporter gene driven by the BACE1 (ß-site amyloid precursor protein cleaving enzyme 1) promoter, which included a portion of the 5' untranslated region (5'UTR). We identified that CL82198, a selective inhibitor of MMP13, decreased BACE1 protein levels in cultured neuronal cells. This effect was dependent on PI3K (phosphatidylinositide 3-kinase) signalling, and was unrelated to BACE1 gene transcription and protein degradation. Further, we found that eukaryotic translation initiation factor 4B (eIF4B) played a key role, as the mutation of eIF4B at serine 422 (S422R) or deletion of the BACE1 5'UTR attenuated MMP13-mediated BACE1 regulation. In APPswe/PS1E9 mice, an animal model of Alzheimer's disease, hippocampal Mmp13 knockdown or intraperitoneal CL82198 administration reduced BACE1 protein levels and the related amyloid-ß precursor protein processing, amyloid-ß load and eIF4B phosphorylation, whereas spatial and associative learning and memory performances were improved. Collectively, MMP13 inhibition/CL82198 treatment exhibited therapeutic potential for Alzheimer's disease, via the translational regulation of BACE1.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Benzofuranos/uso terapéutico , Disfunción Cognitiva/tratamiento farmacológico , Metaloproteinasa 13 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/uso terapéutico , Morfolinas/uso terapéutico , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Células Cultivadas , Factores Eucarióticos de Iniciación/genética , Técnicas de Silenciamiento del Gen , Hipocampo/metabolismo , Humanos , Ratones , Ratones Transgénicos , Mutación , Oligopéptidos/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas
5.
Exp Ther Med ; 14(6): 5751-5756, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29285117

RESUMEN

Myc-interacting zinc-finger protein 1 (Miz1) is a member of the poxvirus and zinc-finger domain/zinc finger transcription factor family. Its transcription activation and repression functions in the nucleus are well elucidated; however its cytoplasmic inflammation function is poorly understood and may be associated with the pathogenesis of Alzheimer's disease (AD). The aim of the present study was to investigate the association between AD and Miz1 expression. In the present study, the expression and distribution of Miz1 in wild-type (WT) and amyloid precursor protein/presenelin-1 (AD) mice was studied using reverse transcription-quantitative polymerase chain reaction, western blot analysis, and immunohistochemical and immunofluorescence staining. The results indicated that Miz1 was significantly upregulated in the cortex of AD mice (P<0.05). Double immunofluorescence labeling revealed that Miz1 protein was predominantly expressed in neurons and astrocytes, as evidenced by co-localization with the dendritic markers microtubule associated protein 2 and glial fibrillary acidic protein, respectively. The results of the present study suggest that the expression of Miz1 in the brain tissue of AD mice may serve an important role in AD pathogenesis.

6.
Oncotarget ; 8(36): 60159-60172, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28947961

RESUMEN

Using high-throughput small molecule screening targeting furin gene, we identified that phorbol esters dPPA (12-Deoxyphorbol 13-phenylacetate 20-acetate) and dPA (12-Deoxyphorbol 13-acetate) significantly increased furin protein and mRNA expression in SH-SY5Y cells. This effect was prevented by PKC (protein kinase C) inhibitor calphostin C but not Ro318220, suggesting that the C1 domain, rather than the catalytic domain of PKC plays an important role. Luciferase assay revealed that nucleotides -7925 to -7426 were sufficient to mediate dPPA/dPA enhancement of furin P1 promoter activity. RNA interference of transcriptional factors CEBPß (CCAAT/enhancer-binding protein ß) and GATA1 revealed that knockdown of CEBPß significantly attenuated the effect of dPPA on furin expression. Pharmacological inhibition of ERK and PI3K but not TGFß receptor diminished the up-regulation of furin by dPPA. These results suggested that in neuronal cells, transcriptional activation of furin by dPPA/dPA may be initiated by C1 domain containing proteins including PKC; the intracellular signaling involves ERK and PI3K and transcription factor CEBPß.

7.
Biochem Biophys Res Commun ; 486(2): 492-498, 2017 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-28320515

RESUMEN

HMGCS2 (mitochondrial 3-hydroxy-3-methylglutaryl-COA synthase 2) is a control enzyme in ketogenesis. The mitochondrial localization and interaction with APP (ß-amyloid precursor protein) suggest that HMGCS2 may play a role in the pathophysiology of AD (Alzheimer's disease). Here we report that overexpression of HMGCS2 decreased levels of APP and related CTFs (carboxy-terminal fragments), which was largely prevented by an autophagic inhibitor chloroquine. In addition, HMGCS2 enhancement of autophagic marker LC3II was diminished by rapamycin, an inhibitor of mechanistic target of rapamycin. Moreover, deprivation of EBSS (Earle's Balanced Salt Solution) significantly augmented the effect of HMGCS2 on LC3II, while acetoacetate reversed the reduction of LC3II, APP and CTFs which was induced by HMGCS2 knockdown. In the presence of acetoacetate, rapamycin failed to induce further increase of LC3II, which mimicked the effect of HMGCS2 overexpression. Finally, HMGCS2 enhanced the antioxidant response. Collectively, HMGCS2 shares with ketone bodies common features in autophagic clearance of APP and CTFs, suggesting that ketone bodies play an important role in HMGCS2 regulation of the autophagy.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Autofagia/genética , Hidroximetilglutaril-CoA Sintasa/genética , Cuerpos Cetónicos/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Serina-Treonina Quinasas TOR/genética , Acetoacetatos/farmacología , Animales , Línea Celular , Cloroquina/farmacología , Regulación de la Expresión Génica , Células HEK293 , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Hidroximetilglutaril-CoA Sintasa/antagonistas & inhibidores , Hidroximetilglutaril-CoA Sintasa/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteolisis/efectos de los fármacos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Transgenes
8.
J Mol Neurosci ; 61(3): 436-448, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28150190

RESUMEN

Estrogen exerts multiple actions in the brain and is an important neuroprotective factor in a number of neuronal disorders. However, the underlying mechanism remains unknown. Studies demonstrate that ubiquitin-conjugating enzyme 9 (ubc9) has an integral role in synaptic plasticity and may contribute to the pathology of neuronal disorders. We aimed to investigate the effects of estrogen on ubc9 and in the Alzheimer's disease brain. Ubc9 protein and mRNA were significantly increased in the cortex and hippocampus of APP/PS1 mice with enhanced SUMOylation. Systemic estrogen administration led to reduced ubc9 expression in ovariectomized APP/PS1 mice and reduced SUMOylation. The inhibition of ubc9 expression by estrogen was found to be dose-dependent in cultured neurons. However, estrogen receptor (ER) antagonist ICI182780 did not block the inhibition of ubc9 expression by estrogen. Furthermore, the reduced expression of ubc9 was not mediated by ERα or ERß agonists alone or in combination, but by the membrane-impermeable ER agonist E2-bovine serum albumin. The activation of the G protein-coupled ER mediated the inhibition of ubc9 expression of estrogen. A phosphoinositide 3-kinase (PI3K) inhibitor, rather than an extracellular signal-regulated kinase inhibitor, blocked the inhibition of ubc9 by estrogen. Estrogen treatment significantly increased the phosphorylation of PI3K, which suggests that activation of the PI3K pathway by estrogen is required for ubc9 regulation. Further, ubc9 interacted with the synaptic proteins post-synaptic density protein 95 (PSD95) and synaptophysin. Estrogen decreased the interaction of ubc9 with post-synaptic PSD95, but increased the interaction of ubc9 with pre-synaptic synaptophysin. These results suggest that a membrane-bound ER might mediate the estrogen inhibition of ubc9 in cortical neurons, where PI3K plays an important role. We also show that ubc9 can interact with synaptic proteins, which are subject to estrogen regulation.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Corteza Cerebral/citología , Estrógenos/farmacología , Neuronas/metabolismo , Sinapsis/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Células Cultivadas , Corteza Cerebral/metabolismo , Homólogo 4 de la Proteína Discs Large , Femenino , Guanilato-Quinasas/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Presenilina-1/genética , Unión Proteica , Transporte de Proteínas , Receptores de Estrógenos/metabolismo , Sinapsis/efectos de los fármacos , Sinaptofisina/metabolismo , Enzimas Ubiquitina-Conjugadoras/genética
9.
BMC Genomics ; 18(1): 194, 2017 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-28219337

RESUMEN

BACKGROUND: Nicotine is known to differentially regulate cortical interneuron and pyramidal neuron activities in the neocortex, while the underlying molecular mechanisms have not been well studied. In this study, RNA-sequencing was performed in acutely isolated cortical somatostatin (Sst)- positive interneurons and pyramidal neurons (Thy1) from mice treated with systemic nicotine for 14 days. We assessed the differentially expressed genes (DEGs) by nicotine in Sst- or Thy1- neurons, respectively, and then compared DEGs between Sst- and Thy1- neurons in the absence and presence of nicotine. RESULTS: In Sst-neurons, the DEGs by nicotine were associated with glycerophospholipid and nicotinate and nicotinamide metabolism; while in Thy1-neurons those related to immune response and purine and pyrimidine metabolisms were affected. Under basal condition, the DEGs between Sst- and Thy1- neurons were frequently associated with signal transduction, phosphorylation and potassium channel regulation. However, some new DEGs between Sst- and Thy1- neurons were found after nicotine, the majority of which belong to mitochondrial respiratory chain complex. CONCLUSIONS: Nicotine differentially affected subset of genes in Sst- and Thy1- neurons, which might contribute to the distinct effect of nicotine on interneuron and pyramidal neuron activities. Meanwhile, the altered transcripts associated with mitochondrial activity were found between interneurons and pyramidal neurons after chronic nicotine.


Asunto(s)
Encéfalo/citología , Perfilación de la Expresión Génica , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Nicotina/farmacología , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Animales , Encéfalo/efectos de los fármacos , Ratones , Análisis de Secuencia de ARN , Factores de Tiempo
10.
Mol Neurobiol ; 54(6): 4113-4126, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-27324789

RESUMEN

Neuroplasticity refers to the changes in the molecular and cellular processes of neural circuits that occur in response to environmental experiences. Clinical and experimental studies have increasingly shown that estrogens participate in the neuroplasticity involved in cognition, behavior, and memory. It is generally accepted that estrogens exert their effects through genomic actions that occur over a period of hours to days. However, emerging evidence indicates that estrogens also rapidly influence the neural circuitry through nongenomic actions. In this review, we provide an overview of the genomic and nongenomic actions of estrogens and discuss how these actions may cooperate in synaptic plasticity. We then summarize the role of epigenetic modifications, synaptic protein synthesis, and posttranslational modifications, and the splice variants of estrogen receptors in the complicated network of estrogens. The combination of genomic and nongenomic mechanisms endows estrogens with considerable diversity in modulating neural functions including synaptic plasticity.


Asunto(s)
Estrógenos/farmacología , Genoma , Plasticidad Neuronal/genética , Animales , Humanos , Modelos Biológicos , Plasticidad Neuronal/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Receptores de Estrógenos/química , Receptores de Estrógenos/metabolismo
11.
FASEB J ; 31(4): 1482-1493, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28003340

RESUMEN

ADAM10 (a disintegrin and metalloproteinase domain-containing protein 10) is the α-secretase that is involved in APP (ß-amyloid precursor protein) processing. Enhancement of the nonamyloidogenic APP pathway by ADAM10 provides therapeutic potential for Alzheimer's disease (AD). By using high-throughput screening that targeted ADAM10, we determined that apicidin-an inhibitor of HDACs (histone deacetylases)-significantly increased mRNA and protein levels of ADAM10 in SH-SY5Y cells. A luciferase assay revealed that the nucleotides -444 to -300 in the ADAM10 promoter were sufficient to mediate this effect. In addition, knockdown of USF1 (upstream transcription factor 1) and HDAC2/3 prevented apicidin regulation of ADAM10. Moreover, USF1 acetylation was increased by apicidin, which enhanced the association of USF1 with HDAC2/3 and with the ADAM10 promoter. We further found that apicidin did not affect the phosphorylation of ERK or USF1; however, ERK inhibitor U0126 blocked the effect of apicidin on ADAM10. Finally, apicidin increased the level of α-site C-terminal fragment from APP and reduced the production of ß-amyloid peptide 1-42. Collectively, our study provides evidence that ADAM10 expression can be regulated by HDAC2/3 inhibitor apicidin via USF1-dependent mechanisms in which ERK signaling plays an important role. Thus, HDAC regulation of ADAM10 might shed new light on the understanding of AD pathology.-Hu, X.-T., Zhu, B.-L., Zhao, L.-G., Wang, J.-W., Liu, L., Lai, Y.-J., He, L., Deng, X.-J., Chen, G.-J. Histone deacetylase inhibitor apicidin increases expression of the α-secretase ADAM10 through transcription factor USF1-mediated mechanisms.


Asunto(s)
Proteína ADAM10/genética , Secretasas de la Proteína Precursora del Amiloide/genética , Inhibidores de Histona Desacetilasas/farmacología , Proteínas de la Membrana/genética , Péptidos Cíclicos/farmacología , Factores Estimuladores hacia 5'/metabolismo , Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Línea Celular Tumoral , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Activación Transcripcional/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Factores Estimuladores hacia 5'/genética
12.
J Mol Neurosci ; 60(2): 205-13, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27498200

RESUMEN

L-type calcium channel (LTCC) gene Cav1.2 is believed to play an important role in the alteration of Ca(2+) homeostasis in brain astrocytes. Increasing evidence shows that alteration of intracellular Ca(2+) concentration is related to the effect of 17ß-estradiol (E2) in a variety of neurophysiological and neuropathological conditions. In this study, we measured immunoreactivity of Cav1.2 protein expression in rat primary cortical astrocytes by using Western blots. We demonstrated that E2 upregulated Cav1.2 expression in a dose- and time-dependent manner and the effect of E2 on Cav1.2 expression were blocked by an estrogen receptor (ER) antagonist, ICI-182,780. The ER subtype-selective ERα agonists propylpyrazole triole (PPT) and ERß agonist diarylpropionitrile (DPN) both increase the expression of Cav1.2 in a dose-dependent manner. Also, the PPT most closely mimicked the upregulation of Cav1.2 protein expression by E2. Similar experiments of 10 nM E2-treated ERα- or ERß-knockdown astrocytes have also shown that the E2 regulation of Cav1.2 protein expression is mediated through an ERα-dependent pathway. Furthermore, we established that E2 did not change the level of Cav1.2 mRNA. The induction of E2-mediated Cav1.2 expression was inhibited by cycloheximide (CHX) but not by actinomycin D (Act-D), suggesting that E2 regulation of Cav1.2 expression occurred at a posttranscriptional level. We also found that E2 may increase Cav1.2 levels by decreasing its ubiquitination and degradation rate. These findings provide new information about the effect of E2 on Cav1.2 in astrocytes, particularly necessary for the treatment of neurological disease.


Asunto(s)
Astrocitos/metabolismo , Canales de Calcio Tipo L/metabolismo , Corteza Cerebral/citología , Estradiol/farmacología , Receptores de Estrógenos/metabolismo , Animales , Astrocitos/efectos de los fármacos , Canales de Calcio Tipo L/genética , Células Cultivadas , Corteza Cerebral/metabolismo , Proteolisis , Ratas , Ratas Sprague-Dawley , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Estrógenos/genética , Ubiquitinación , Regulación hacia Arriba
13.
Genes Dis ; 2(4): 291-292, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30258871
14.
Pediatr Neonatol ; 49(5): 189-92, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19133571

RESUMEN

We report on the clinical, cytogenetic, and imaging findings in a patient with a 7q terminal deletion. The 11-year-old girl had mental retardation, microcephaly, a distinctive face, relatively small hands and feet, and sacral dysgenesis. High resolution GTG banding (550-850 bands) showed a 7q terminal deletion. A detailed evaluation of associated malformations and the overall clinical picture should be taken into account when identifying the underlying diagnosis in cases of sacral dysgenesis with mental retardation.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 7 , Sacro/anomalías , Anomalías Múltiples , Niño , Femenino , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...