Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Eur J Pharmacol ; 969: 176452, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38417609

RESUMEN

Seeking an effective and safe scheme is the common goal of clinical treatment of tumor patients. In recent years, traditional Chinese medicine has attracted more and more attention in order to discover new drugs with good anti-tumor effects. Oroxylin A (OA) is a compound found in natural Oroxylum indicum and Scutellaria baicalensis Georgi plants and has been used in the treatment of various cancers. Studies have shown that OA has a wide range of powerful biological activities and plays an important role in neuroprotection, anti-inflammation, anti-virus, anti-allergy, anti-tumor and so on. OA shows high efficacy in tumor treatment. Therefore, it has attracted great attention of researchers all over the world. This review aims to discuss the anti-tumor effects of OA from the aspects of cell cycle arrest, induction of cell proliferation and apoptosis, induction of autophagy, anti-inflammation, inhibition of glycolysis, angiogenesis, invasion, metastasis and reversal of drug resistance. In addition, the safety and toxicity of the compound were also discussed. As a next step, to clarify the benefits and adverse effects of Oroxylin A in cancer patients further experiments, especially clinical trials, are needed.


Asunto(s)
Flavonoides , Neoplasias , Humanos , Flavonoides/farmacología , Flavonoides/uso terapéutico , Apoptosis , Proliferación Celular , Autofagia , Neoplasias/tratamiento farmacológico
2.
Sci Total Environ ; 884: 163748, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37120017

RESUMEN

BACKGROUND: Evidence of the association between particles with a diameter of 2.5 µm or less (PM2.5) in long term and ovarian cancer (OC) mortality is limited. METHODS: This prospective cohort study analyzed data collected between 2015 and 2020 from 610 newly diagnosed OC patients, aged 18-79 years. The residential average PM2.5 concentrations 10 years before the date of OC diagnosis were assessed by random forest models at a 1 km × 1 km resolution. Cox proportional hazard models fully adjusted for the covariates (including age at diagnosis, education, physical activity, kitchen ventilation, FIGO stage, and comorbidities) and distributed lag non-linear models were used to estimate the hazard ratios (HRs) and 95 % confidence intervals (CIs) of PM2.5 and all-cause mortality of OC. RESULTS: During a median follow-up of 37.6 months (interquartile: 24.8-50.5 months), 118 (19.34 %) deaths were confirmed among 610 OC patients. One-year PM2.5 exposure levels before OC diagnosis was significantly associated with an increase in all-cause mortality among OC patients (single-pollutant model: HR = 1.22, 95 % CI: 1.02-1.46; multi-pollutant models: HR = 1.38, 95 % CI: 1.10-1.72). Furthermore, during 1 to 10 years prior to diagnosis, the lag-specific effect of long-term PM2.5 exposure on the all-cause mortality of OC had a risk increase for lag 1-6 years, and the exposure-response relationship was linear. Of note, significant interactions between several immunological indicators as well as solid fuel use for cooking and ambient PM2.5 concentrations were observed. CONCLUSION: Higher ambient PM2.5 concentrations were associated with an increased risk of all-cause mortality among OC patients, and there was a lag effect in long-term PM2.5 exposure.


Asunto(s)
Contaminantes Atmosféricos , Contaminación del Aire , Contaminantes Ambientales , Neoplasias Ováricas , Humanos , Femenino , Material Particulado/efectos adversos , Contaminantes Atmosféricos/análisis , Estudios Prospectivos , Exposición a Riesgos Ambientales/efectos adversos
3.
Yi Chuan ; 44(11): 993-1008, 2022 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-36384993

RESUMEN

Prime editing is a newly developed CRISPR/Cas system-based genome editing technique. The effector of prime editor (PE) is termed PE2, which is generated by fusing a reverse transcriptase (RT) with a Cas9 H840A nickase. The guide RNA of PE is termed prime editing guide RNA (pegRNA), which consists of a single guide RNA (sgRNA) with a 3' extension containing the RT template (RTT) and primer binding site (PBS). PE can install all 12 types of point mutations, small insertions and deletions and combinations thereof. Since its emergence in 2019, with the high versatility and specificity, PE has been applied to many living organisms, including animals, plants and bacteria. This led to many explorations of PE on gene therapy and genetic improvement in agriculture. In this review, we systematically describe the development, characteristics, optimizations, applications and security of PE. In addition, we discuss the future applications of PE. We expect that this review will help researchers to grasp and better use PE.


Asunto(s)
Edición Génica , ARN Pequeño no Traducido , Animales , Sistemas CRISPR-Cas/genética , Edición Génica/métodos , Plantas/genética , Mutación Puntual , ARN Pequeño no Traducido/genética
4.
Front Mol Neurosci ; 15: 1014251, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36267702

RESUMEN

Neurodegenerative diseases are a class of incurable and debilitating diseases characterized by progressive degeneration and death of cells in the central nervous system. They have multiple underlying mechanisms; however, they all share common degenerative features, such as mitochondrial dysfunction. According to recent studies, neurodegenerative diseases are associated with the accumulation of dysfunctional mitochondria. Selective autophagy of mitochondria, called mitophagy, can specifically degrade excess or dysfunctional mitochondria within cells. In this review, we highlight recent findings on the role of mitophagy in neurodegenerative disorders. Multiple studies were collected, including those related to the importance of mitochondria, the mechanism of mitophagy in protecting mitochondrial health, and canonical and non-canonical pathways in mitophagy. This review elucidated the important function of mitophagy in neurodegenerative diseases, discussed the research progress of mitophagy in neurodegenerative diseases, and summarized the role of mitophagy-related proteins in neurological diseases. In addition, we also highlight pharmacological advances in neurodegeneration.

5.
J Biol Chem ; 298(6): 102054, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35598826

RESUMEN

Myosins belong to a large superfamily of actin-dependent molecular motors. Nonmuscle myosin II (NM II) is involved in the morphology and function of neurons, but little is known about how NM II activity is regulated. Brain-derived neurotrophic factor (BDNF) is a prevalent neurotrophic factor in the brain that encourages growth and differentiation of neurons and synapses. In this study, we report that BDNF upregulates the phosphorylation of myosin regulatory light chain (MLC2), to increases the activity of NM II. The role of BDNF on modulating the phosphorylation of MLC2 was validated by using Western blotting in primary cultured hippocampal neurons. This result was confirmed by injecting BDNF into the dorsal hippocampus of mice and detecting the phosphorylation level of MLC2 by Western blotting. We further perform coimmunoprecipitation assay to confirm that this process depends on the activation of the LYN kinase through binding with tyrosine kinase receptor B, the receptor of BDNF, in a kinase activity-dependent manner. LYN kinase subsequently phosphorylates MLCK, further promoting the phosphorylation of MLC2. Taken together, our results suggest a new molecular mechanism by which BDNF regulates MLC2 activity, which provides a new perspective for further understanding the functional regulation of NM II in the nervous system.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Cadenas Ligeras de Miosina , Miosina Tipo II , Quinasa de Cadena Ligera de Miosina , Familia-src Quinasas , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Células Cultivadas , Hipocampo/metabolismo , Ratones , Cadenas Ligeras de Miosina/metabolismo , Miosina Tipo II/metabolismo , Quinasa de Cadena Ligera de Miosina/química , Neuronas/metabolismo , Fosforilación , Familia-src Quinasas/metabolismo
6.
Technol Cancer Res Treat ; 21: 15330338221082431, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35243940

RESUMEN

Background: Malignant melanoma is a common malignant tumor and one of the tumors with the fastest growing incidence. The effect of microRNAs on the biological processing of malignant melanoma cells also have been reported. This study explores the ability of miR-498 to regulate the progression of malignant melanoma cells. Methods: The expression of miR-498 was detected by RT-qPCR. The proliferation, invasion, and migration of malignant melanoma cells were measured by cell counting kit-8, clone formation, and transwell assays. Flow cytometry assay detected the percentage of apoptotic cells. Western blot was used to detect the expression of markers related to epithelial-mesenchymal transition. The correction of miR-498 and UBE2T was explored by dual-luciferase assay and Western blot. Results: Overexpression of miR-498 inhibited the proliferation, invasion, migration, and induced cell apoptosis of M14 and A375 cells. In addition, the expression of epithelial-mesenchymal transition-related factors was altered by the overexpression of miR-498. miR-498 can directly target UBE2T 3'-UTR and inhibit UBE2T protein expression. The overexpression of UBE2T reversed the inhibitory effects of miR-498 on the progression of malignant melanoma cells. Furthermore, UBE2T mRNA was significantly highly expressed in malignant melanoma tissues. The high expression of UBE2T was associated with the poor overall survival rate of malignant melanoma patients. Conclusions: Altogether, our findings demonstrated that miR-498 significantly inhibited the proliferation, invasion, migration, and induced apoptosis of malignant melanoma cells and confirmed that miR-498 regulated malignant melanoma cell progression by targeting UBE2T.


Asunto(s)
Melanoma , MicroARNs , Enzimas Ubiquitina-Conjugadoras , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias Cutáneas , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Melanoma Cutáneo Maligno
7.
Front Cell Dev Biol ; 9: 662763, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34485271

RESUMEN

The neuron derived synaptic adhesion molecular neuroligin-3 (NLGN3) plays an important role in glioma growth. While the role of autocrine NLGN3 in glioma has not been well-studied. The expression of NLGN3 in glioma was detected using immunohistochemistry. We further explored its function and regulatory mechanism in U251 and U87 cells with high expression of NLGN3. Knockdown of endogenous NLGN3 significantly reduced the proliferation, migration, and invasion of glioma cells and down-regulated the activity of the PI3K-AKT, ERK1/2, and LYN signaling pathways. In comparison, overexpression of NLGN3 yielded opposite results. Our results further demonstrate that LYN functions as a feedback mechanism to promote NLGN3 cleavage. This feedback regulation was achieved by upregulating the ADAM10 sheddase responsible for NLGN3 cleavage. Inhibition of ADAM10 suppressed the proliferation, migration, and invasion of glioma cells; oppositely, the expression of ADAM10 was correlated with a higher likelihood of lower grade glioma (LGG) in the brain. Our study demonstrates that glioma-derived NLGN3 promotes glioma progression by upregulating activity of LYN and ADAM10, which in turn promote NLGN3 cleavage to form a positive feedback loop. This pathway may open a potential therapeutic window for the treatment of human glioma.

8.
Brain Res ; 1737: 146802, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32220534

RESUMEN

Growth differentiation factor 11 (GDF11), a member of the transforming growth factor-ß (TGF-ß) superfamily, regulates various biological processes in mammals. The effect of GDF11 in brain injury has not been fully elucidated. Our aim was to investigate the effects of GDF11 in cerebral ischemic injury. The expression level of GDF11 increased significantly in the peri-infarct cerebral cortex. Next, the effect of the intracerebroventricular injection of a GDF11 overexpression lentivirus or rGDF11 was investigated in middle cerebral artery occlusion (MCAO) rats. The preventative effects of the GDF11 overexpression virus on stroke were observed. The delivery of the lentivirus into rats before MCAO significantly reduced the infarct volume and the percentage of apoptotic cells and improved motor function in MCAO rats. Furthermore, it elevated the expression of p-Smad2/3 and promoted neurogenesis and angiogenesis in the ipsilateral SVZ during ischemic injury. More importantly, the therapeutic effects of rGDF11 on stroke were subsequently explored. The results in MCAO rats treated with rGDF11 were found similar to that in those treated with the GDF11 overexpression lentivirus. Together, these findings indicate that GDF11 has neuroprotective and neurorestorative effects in cerebral ischemic injury and provide new insights into the function and mechanism of GDF11 in stroke models.


Asunto(s)
Isquemia Encefálica/metabolismo , Factores de Diferenciación de Crecimiento/metabolismo , Animales , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/fisiopatología , Corteza Cerebral , Modelos Animales de Enfermedad , Factores de Diferenciación de Crecimiento/fisiología , Factores de Diferenciación de Crecimiento/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Masculino , Neovascularización Patológica/tratamiento farmacológico , Neurogénesis , Neuroprotección , Fármacos Neuroprotectores , Ratas , Accidente Cerebrovascular/tratamiento farmacológico
9.
Int J Mol Sci ; 21(4)2020 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-32070035

RESUMEN

Cadherin epidermal growth factor (EGF) laminin G (LAG) seven-pass G-type receptor 1 (CELSR1) is a member of a special subgroup of adhesion G protein-coupled receptors. Although Celsr1 has been reported to be a sensitive gene for stroke, the effect of CELSR1 in ischemic stroke is still not known. Here, we investigated the effect of CELSR1 on neuroprotection, neurogenesis and angiogenesis in middle cerebral artery occlusion (MCAO) rats. The mRNA expression of Celsr1 was upregulated in the subventricular zone (SVZ), hippocampus and ischemic penumbra after cerebral ischemic injury. Knocking down the expression of Celsr1 in the SVZ with a lentivirus significantly reduced the proliferation of neuroblasts, the number of CD31-positive cells, motor function and rat survival and increased cell apoptosis and the infarct volume in MCAO rats. In addition, the expression of p-PKC in the SVZ and peri-infarct tissue was downregulated after ischemia/ reperfusion. Meanwhile, in the dentate gyrus of the hippocampus, knocking down the expression of Celsr1 significantly reduced the proliferation of neuroblasts; however, it had no influence on motor function, cell apoptosis or angiogenesis. These data indicate that CELSR1 has a neuroprotective effect on cerebral ischemia injury by reducing cell apoptosis in the peri-infarct cerebral cortex and promoting neurogenesis and angiogenesis, mainly through the Wnt/PKC pathway.


Asunto(s)
Isquemia Encefálica/genética , Cadherinas/genética , Neurogénesis/genética , Accidente Cerebrovascular/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Isquemia Encefálica/patología , Proliferación Celular/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/patología , Ventrículos Laterales/metabolismo , Ventrículos Laterales/patología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Fármacos Neuroprotectores/metabolismo , ARN Mensajero/genética , Ratas , Accidente Cerebrovascular/patología , Vía de Señalización Wnt/genética
10.
Int J Mol Sci ; 19(7)2018 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-29966219

RESUMEN

Cerebral dopamine neurotrophic factor (CDNF), previously known as the conserved dopamine neurotrophic factor, belongs to the evolutionarily conserved CDNF/mesencephalic astrocyte-derived neurotrophic factor MANF family of neurotrophic factors that demonstrate neurotrophic activities in dopaminergic neurons. The function of CDNF during brain ischemia is still not known. MANF is identified as an endoplasmic reticulum (ER) stress protein; however, the role of CDNF in ER stress remains to be fully elucidated. Here, we test the neuroprotective effect of CDNF on middle cerebral artery occlusion (MCAO) rats and neurons and astrocytes treated with oxygen⁻glucose depletion (OGD). We also investigate the expression of CDNF in cerebral ischemia and in primary neurons treated with ER stress-inducing agents. Our results show that CDNF can significantly reduce infarct volume, reduce apoptotic cells and improve motor function in MCAO rats, while CDNF can increase the cell viability of neurons and astrocytes treated by OGD. The expression of CDNF was upregulated in the peri-infarct tissue at 2 h of ischemia/24 h reperfusion. ER stress inducer can induce CDNF expression in primary cultured neurons. Our data indicate that CDNF has neuroprotective effects on cerebral ischemia and the OGD cell model and the protective mechanism of CDNF may occur through ER stress pathways.


Asunto(s)
Isquemia Encefálica/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Factores de Crecimiento Nervioso/metabolismo , Animales , Western Blotting , Isquemia Encefálica/genética , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Células Cultivadas , Estrés del Retículo Endoplásmico/genética , Glucosa/deficiencia , Inmunohistoquímica , Infarto de la Arteria Cerebral Media/metabolismo , Masculino , Factores de Crecimiento Nervioso/genética , Oxígeno/metabolismo , Ratas , Ratas Sprague-Dawley
11.
Analyst ; 143(6): 1367-1377, 2018 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-29423467

RESUMEN

In this study, we developed an automated microfluidic DNA microarray (AMDM) platform for point mutation detection of genetic variants in inherited arrhythmic diseases. The platform allows for automated and programmable reagent sequencing under precise conditions of hybridization flow and temperature control. It is composed of a commercial microfluidic control system, a microfluidic microarray device, and a temperature control unit. The automated and rapid hybridization process can be performed in the AMDM platform using Cy3 labeled oligonucleotide exons of SCN5A genetic DNA, which produces proteins associated with sodium channels abundant in the heart (cardiac) muscle cells. We then introduce a graphene oxide (GO)-assisted DNA microarray hybridization protocol to enable point mutation detection. In this protocol, a GO solution is added after the staining step to quench dyes bound to single-stranded DNA or non-perfectly matched DNA, which can improve point mutation specificity. As proof-of-concept we extracted the wild-type and mutant of exon 12 and exon 17 of SCN5A genetic DNA from patients with long QT syndrome or Brugada syndrome by touchdown PCR and performed a successful point mutation discrimination in the AMDM platform. Overall, the AMDM platform can greatly reduce laborious and time-consuming hybridization steps and prevent potential contamination. Furthermore, by introducing the reciprocating flow into the microchannel during the hybridization process, the total assay time can be reduced to 3 hours, which is 6 times faster than the conventional DNA microarray. Given the automatic assay operation, shorter assay time, and high point mutation discrimination, we believe that the AMDM platform has potential for low-cost, rapid and sensitive genetic testing in a simple and user-friendly manner, which may benefit gene screening in medical practice.


Asunto(s)
Arritmias Cardíacas/genética , Técnicas Analíticas Microfluídicas , Análisis de Secuencia por Matrices de Oligonucleótidos , Arritmias Cardíacas/diagnóstico , Análisis Mutacional de ADN , Humanos , Canal de Sodio Activado por Voltaje NAV1.5/genética , Hibridación de Ácido Nucleico , Oligonucleótidos , Mutación Puntual
12.
J Neurosci ; 37(25): 5978-5995, 2017 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-28500221

RESUMEN

Multiple studies have established that brain-derived neurotrophic factor (BDNF) plays a critical role in the regulation of synaptic plasticity via its receptor, TrkB. In addition to being phosphorylated, TrkB has also been demonstrated to be ubiquitinated. However, the mechanisms of TrkB ubiquitination and its biological functions remain poorly understood. In this study, we demonstrate that ubiquitin C-terminal hydrolase L1 (UCH-L1) promotes contextual fear conditioning learning and memory via the regulation of ubiquitination of TrkB. We provide evidence that UCH-L1 can deubiquitinate TrkB directly. K460 in the juxtamembane domain of TrkB is the primary ubiquitination site and is regulated by UCH-L1. By using a peptide that competitively inhibits the association between UCH-L1 and TrkB, we show that the blockade of UCH-L1-regulated TrkB deubiquitination leads to increased BDNF-induced TrkB internalization and consequently directs the internalized TrkB to the degradation pathway, resulting in increased degradation of surface TrkB and attenuation of TrkB activation and its downstream signaling pathways. Moreover, injection of the peptide into the DG region of mice impairs hippocampus-dependent memory. Together, our results suggest that the ubiquitination of TrkB is a mechanism that controls its downstream signaling pathways via the regulation of its endocytosis and postendocytic trafficking and that UCH-L1 mediates the deubiquitination of TrkB and could be a potential target for the modulation of hippocampus-dependent memory.SIGNIFICANCE STATEMENT Ubiquitin C-terminal hydrolase L1 (UCH-L1) has been demonstrated to play important roles in the regulation of synaptic plasticity and learning and memory. TrkB, the receptor for brain-derived neurotrophic factor, has also been shown to be a potent regulator of synaptic plasticity. In this study, we demonstrate that UCH-L1 functions as a deubiquitinase for TrkB. The blockage of UCH-L1-regulated deubiquitination of TrkB eventually results in the increased degradation of surface TrkB and decreased activation of TrkB and its downstream signaling pathways. In vivo, UCH-L1-regulated TrkB deubiquitination is necessary for hippocampus-dependent memory. Overall, our study provides novel insights into the mechanisms of UCH-L1-mediated neurobiological functions and suggests that ubiquitination is an important regulatory signal for TrkB functions.


Asunto(s)
Hipocampo/fisiología , Memoria/fisiología , Receptor trkB/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación/fisiología , Animales , Azepinas/farmacología , Benzamidas/farmacología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/farmacología , Condicionamiento Operante/fisiología , Endocitosis/genética , Endocitosis/fisiología , Miedo/psicología , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Actividad Motora/fisiología , Neuronas/metabolismo , Receptor trkB/antagonistas & inhibidores , Transducción de Señal/genética , Transducción de Señal/fisiología , Ubiquitina Tiolesterasa/genética , Ubiquitinación/genética
13.
Oncotarget ; 8(64): 108238-108248, 2017 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-29296237

RESUMEN

MARCH8 belongs to a family of membrane-associated RING-CH (MARCH) ubiquitin ligases. The functions of MARCH8 have been thoroughly investigated but its mechanism of action remains unknown. In this study, we detected the expression of MARCH8 protein in NSCLC samples and identified MARCH8 mRNA expression through a TCGA database. In addition, we analyzed the correlation between MARCH8 and the clinical characteristics of NSCLC patients and their prognosis.(www.kmplot.com). The roles of MARCH8 in proliferation, migration, and metastasis were further explored through ectopic expression analysis and western blot analysis; its mechanism of expressionwas also explored. We discovered that MARCH8 was downregulated in NSCLC tissues compared to adjacent normal lung tissues. Overexpression of MARCH8 inhibited NSCLC cell proliferation and metastasis via the PI3K and mTOR signaling pathways; this also increased apoptosis of A549 and H1299 cells. Our results indicated that MARCH8 plays crucial roles in NSCLC against carcinogenesis and progression; therefore, MARCH8 might be a predictive factor and an attractive therapeutic target for NSCLC patients.

14.
J Neurochem ; 135(3): 453-65, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26303065

RESUMEN

Brain-derived neurotrophic factor (BDNF) has been implicated in the potent modulation of synaptic plasticity at both pre-synaptic and post-synaptic sites. However, the molecular mechanism underlying BDNF-mediated pre-synaptic modulation remains incompletely understood. Here, we report that BDNF treatment for over 4 h could significantly enhance the expression of c-Jun NH2-terminal kinase-interacting protein 3 (JIP3) in cultured hippocampal neurons. This enhancement could be blocked by the Trk inhibitor K252a or by a cAMP response element-binding protein (CREB) inhibitor. In addition, chromatin immunoprecipitation (ChIP) assays revealed that CREB could bind with the JIP3 promoter region and the BDNF treatment could increase this binding. Using dual-luciferase assays we further characterized the cAMP response element (CRE) site in the JIP3 promoter. Finally, we found that BDNF-increased JIP3 expression contributes to the BDNF-induced modulation of neurotransmitter release. Together, our studies reveal that in hippocampal neurons BDNF up-regulates JIP3 expression via CREB activation, which contributes to the enhancement of neurotransmitter release; thus, we have identified a novel mechanism that BDNF modulates pre-synaptic transmission.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Proteína Quinasa 10 Activada por Mitógenos/biosíntesis , Regulación hacia Arriba/fisiología , Animales , Proteína de Unión a CREB/metabolismo , Células Cultivadas , Células HEK293 , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Ratones , Proteína Quinasa 10 Activada por Mitógenos/metabolismo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos
15.
Dev Cell ; 33(6): 690-702, 2015 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-26004511

RESUMEN

Recent studies in humans and in genetic mouse models have identified Slit- and NTRK-like family (Slitrks) as candidate genes for neuropsychiatric disorders. All Slitrk isotypes are highly expressed in the CNS, where they mediate neurite outgrowth, synaptogenesis, and neuronal survival. However, the molecular mechanisms underlying these functions are not known. Here, we report that Slitrk5 modulates brain-derived neurotrophic factor (BDNF)-dependent biological responses through direct interaction with TrkB receptors. Under basal conditions, Slitrk5 interacts primarily with a transsynaptic binding partner, protein tyrosine phosphatase δ (PTPδ); however, upon BDNF stimulation, Slitrk5 shifts to cis-interactions with TrkB. In the absence of Slitrk5, TrkB has a reduced rate of ligand-dependent recycling and altered responsiveness to BDNF treatment. Structured illumination microscopy revealed that Slitrk5 mediates optimal targeting of TrkB receptors to Rab11-positive recycling endosomes through recruitment of a Rab11 effector protein, Rab11-FIP3. Thus, Slitrk5 acts as a TrkB co-receptor that mediates its BDNF-dependent trafficking and signaling.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptor trkB/metabolismo , Animales , Cuerpo Estriado/metabolismo , Endosomas/metabolismo , Células HEK293 , Humanos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Unión Proteica , Transporte de Proteínas , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rab/metabolismo
16.
J Cell Sci ; 128(6): 1108-22, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25632160

RESUMEN

Brain-derived neurotrophic factor (BDNF) plays an important role in neuronal survival, neurite outgrowth and synaptic plasticity by activating the receptor tropomyosin receptor kinase B (TrkB, also known as NTRK2). TrkB has been shown to undergo recycling after BDNF stimulation. We have previously reported that full-length TrkB (TrkB-FL) are recycled through a Rab11-dependent pathway upon BDNF stimuli, which is important for the translocation of TrkB-FL into dendritic spines and for the maintenance of prolonged BDNF downstream signaling during long-term potentiation (LTP). However, the identity of the motor protein that mediates the local transfer of recycled TrkB-FL back to the plasma membrane remains unclear. Here, we report that the F-actin-based motor protein myosin Va (Myo5a) mediates the postendocytic recycling of TrkB-FL. Blocking the interaction between Rab11 and Myo5a by use of a TAT-tagged peptide consisting of amino acids 55-66 of the Myo5a ExonE domain weakened the association between TrkB-FL and Myo5a and thus impaired TrkB-FL recycling and BDNF-induced TrkB-FL translocation into dendritic spines. Finally, inhibiting Myo5a-mediated TrkB-FL recycling led to a significant reduction in prolonged BDNF downstream signaling. Taken together, these results show that Myo5a mediates BDNF-dependent TrkB-FL recycling and contributes to BDNF-induced TrkB spine translocation and prolonged downstream signaling.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Espinas Dendríticas/metabolismo , Endocitosis/fisiología , Hipocampo/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo V/metabolismo , Neuronas/metabolismo , Receptor trkB/metabolismo , Animales , Western Blotting , Factor Neurotrófico Derivado del Encéfalo/genética , Células Cultivadas , Hipocampo/citología , Potenciación a Largo Plazo , Espectrometría de Masas , Ratones , Cadenas Pesadas de Miosina/genética , Miosina Tipo V/genética , Plasticidad Neuronal , Neuronas/citología , Transporte de Proteínas , Ratas , Receptor trkB/genética , Transducción de Señal
17.
Cereb Cortex ; 25(11): 4062-75, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24904070

RESUMEN

The Wnt signaling pathway plays critical roles in development. However, to date, the role of Wnts in learning and memory in adults is still not well understood. Here, we aimed to investigate the roles and mechanisms of Wnts in hippocampal-dependent contextual fear conditioning (CFC) memory formation in adult mice. CFC training induced the secretion and expression of Wnt3a and the activation of its downstream Wnt/Ca(2+) and Wnt/ß-catenin signaling pathways in the dorsal hippocampus (DH). Intrahippocampal infusion of Wnt3a antibody impaired CFC acquisition and consolidation, but not expression. Using the Wnt antagonist sFRP1 or the canonical Wnt inhibitor Dkk1, we found that Wnt/Ca(2+) and Wnt/ß-catenin signaling pathways were involved in acquisition and consolidation, respectively. Moreover, we found Wnt3a signaling is not only necessary but also sufficient for CFC memory. Intrahippocampal infusion of exogenous Wnt3a could enhance acquisition and consolidation of CFC. Overexpression of constitutively active ß-catenin in the DH could rescue the deficit in CFC memory consolidation, but not acquisition induced by Wnt3a antibody injection, which suggests ß-catenin signaling pathway acts downstream of Wnt3a to mediate CFC memory consolidation. Our study may help further the understanding of the precise regulation of Wnt3a in differential memory phases depending on divergent signaling pathways.


Asunto(s)
Condicionamiento Psicológico/fisiología , Miedo/psicología , Hipocampo/metabolismo , Memoria/fisiología , Vía de Señalización Wnt/fisiología , Proteína Wnt3/metabolismo , Análisis de Varianza , Animales , Anticuerpos/farmacología , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Condicionamiento Psicológico/efectos de los fármacos , Miedo/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Transducción Genética , Vía de Señalización Wnt/efectos de los fármacos , Proteína Wnt3/genética , Proteína Wnt3/inmunología , beta Catenina
18.
J Neurosci ; 33(21): 9214-30, 2013 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-23699532

RESUMEN

Brain-derived neurotrophic factor (BDNF) plays an important role in the activity-dependent regulation of synaptic structure and function via tropomyosin related kinase B (TrkB) receptor activation. However, whether BDNF could regulate TrkB levels at synapse during long-term potentiation (LTP) is still unknown. We show in cultured rat hippocampal neurons that chemical LTP (cLTP) stimuli selectively promote endocytic recycling of BDNF-dependent full-length TrkB (TrkB-FL) receptors, but not isoform T1 (TrkB.T1) receptors, via a Rab11-dependent pathway. Moreover, neuronal-activity-enhanced TrkB-FL recycling could facilitate receptor translocation to postsynaptic density and enhance BDNF-induced extracellular signal-regulated kinase and phosphatidylinositol 3-kinase activation and rat hippocampal neuron survival. Finally, we found that cLTP could stimulate the switch of Rab11 from an inactive to an active form and that GTP-bound Rab11 could enhance the interaction between TrkB-FL and PSD-95. Therefore, the recycling endosome could serve as a reserve pool to supply TrkB-FL receptors for LTP maintenance. These findings provide a mechanistic link between Rab11-dependent endocytic recycling and TrkB modulation of synaptic plasticity.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Potenciación a Largo Plazo/fisiología , Neuronas/fisiología , Densidad Postsináptica/fisiología , Receptor trkB/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Análisis de Varianza , Animales , Animales Recién Nacidos , Biotinilación , Homólogo 4 de la Proteína Discs Large , Estimulación Eléctrica , Embrión de Mamíferos , Femenino , Hipocampo/citología , Inmunoprecipitación , Etiquetado Corte-Fin in Situ , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Proteínas de la Membrana/metabolismo , Neuronas/ultraestructura , Técnicas de Placa-Clamp , Densidad Postsináptica/genética , Unión Proteica , Transporte de Proteínas/fisiología , Ratas , Ratas Sprague-Dawley , Fracciones Subcelulares/metabolismo , Proteínas de Unión al GTP rab/genética
19.
Mol Med Rep ; 6(6): 1276-80, 2012 12.
Artículo en Inglés | MEDLINE | ID: mdl-23007279

RESUMEN

The novel member of the Rab family of GTPases, Rab23, is an essential negative regulator of the Sonic hedgehog (Shh) signaling pathway. Loss of function mutation of the Rab23 gene causes abnormal development of the neural tube in mice and in certain human congenital diseases. The aberrant overexpression of Rab23 has been associated with various diseases, such as gastric, hepatocellular and lung cancer. The exact function of Rab23 in hepatocellular carcinomas (HCCs), however, remains unknown. Previously, we reported the abnormal sublocalization of Rab23 in lung cancers. In the current study, we investigated the role of Rab23 in HCCs. We report the distinct sublocalization pattern of Rab23 in HCC cell lines. This difference depends on the GDP/GTP-binding form, and inhibition of the Rab23 cycle decreases the expression and nuclear localization of Gli1.


Asunto(s)
Proteínas Hedgehog/metabolismo , Proteínas de Unión al GTP rab/análisis , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Núcleo Celular/metabolismo , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Microscopía Confocal , Mutación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína con Dedos de Zinc GLI1 , Proteínas de Unión al GTP rab/antagonistas & inhibidores , Proteínas de Unión al GTP rab/metabolismo
20.
J Biol Chem ; 287(3): 1932-45, 2012 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-22128160

RESUMEN

The RET tyrosine kinase receptor plays an important role in the development and maintenance of the nervous system. Although the ligand-induced RET signaling pathway has been well described, little is known about the regulation of RET surface expression, which is integral to the cell ability to control the response to ligand stimuli. We found that in dorsal root ganglion (DRG) neurons, which co-express RET and TrkB, the receptor surface levels of RET are significantly higher than that of TrkB. Using a sequence substitution strategy, we identified a key motif (Box1), which is necessary and sufficient for the differential RET and TrkB surface levels. Furthermore, pharmacological and mutagenesis assays revealed that protein kinase C (PKC) and high K(+) depolarization increase RET surface levels through phosphorylation of the Thr(675) residue in the Box1 motif. Finally, we found that the phosphorylation status of the Thr(675) residue influences RET mediated response to GDNF stimulation. In all, these findings provide a novel mechanism for the modulation of RET surface expression.


Asunto(s)
Ganglios Espinales/enzimología , Regulación Enzimológica de la Expresión Génica/fisiología , Neuronas/enzimología , Proteínas Proto-Oncogénicas c-ret/biosíntesis , Receptor trkB/metabolismo , Transducción de Señal/fisiología , Secuencias de Aminoácidos , Animales , Membrana Celular/enzimología , Membrana Celular/genética , Ganglios Espinales/citología , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Neuronas/citología , Células PC12 , Fosforilación/fisiología , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Transporte de Proteínas/fisiología , Proteínas Proto-Oncogénicas c-ret/genética , Ratas , Ratas Sprague-Dawley , Receptor trkB/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA