Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
1.
Small ; 19(29): e2300395, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37029709

RESUMEN

Phenytoin (PHT) is a first-line antiepileptic drug in clinics, which could decrease neuronal bioelectric activity by blocking the voltage-operated sodium channels. However, the intrinsically low blood-brain-barrier (BBB)-crossing capability of PHT and upregulated expression level of the efflux transporter p-glycoprotein (P-gp) coded by the gene Abcb1 in epileptic neurons limit its efficacy in vivo. Herein, a nanointegrated strategy to overcome PHT resistance mechanisms for enhanced antiepileptic efficacy is reported. Specifically, PHT is first incorporated into calcium phosphate (CaP) nanoparticles through biomineralization, followed by the surface modification of the PEGylated BBB-penetrating TAT peptide. The CaP@PHT-PEG-TAT nanoformulation could effectively cross the BBB to be taken in by epileptic neurons. Afterward, the acidic lysosomal environment would trigger their complete degradation to release Ca2+ and PHT into the cytosol. Ca2+ ions would inhibit mitochondrial oxidative phosphorylation to reverse cellular hypoxia to block hypoxia-inducible factor-1α (Hif1α)-Abcb1-axis, as well as disrupt adenosine triphosphate generation, leading to simultaneous suppression of the expression and drug efflux capacity of P-gp to enhance PHT retention. This study offers an approach for effective therapeutic intervention against drug-resistant epilepsy.


Asunto(s)
Epilepsia , Fenitoína , Humanos , Fenitoína/farmacología , Fenitoína/uso terapéutico , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Epilepsia/tratamiento farmacológico , Convulsiones/tratamiento farmacológico , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Neuronas/metabolismo , Fosfatos de Calcio
2.
BMC Genomics ; 23(1): 430, 2022 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-35676651

RESUMEN

BACKGROUND: Seizures are a common symptom in glioma patients, and they can cause brain dysfunction. However, the mechanism by which glioma-related epilepsy (GRE) causes alterations in brain networks remains elusive. OBJECTIVE: To investigate the potential pathogenic mechanism of GRE by analyzing the dynamic expression profiles of microRNA/ mRNA/ lncRNA in brain tissues of glioma patients. METHODS: Brain tissues of 16 patients with GRE and 9 patients with glioma without epilepsy (GNE) were collected. The total RNA was dephosphorylated, labeled, and hybridized to the Agilent Human miRNA Microarray, Release 19.0, 8 × 60 K. The cDNA was labeled and hybridized to the Agilent LncRNA + mRNA Human Gene Expression Microarray V3.0, 4 × 180 K. The raw data was extracted from hybridized images using Agilent Feature Extraction, and quantile normalization was performed using the Agilent GeneSpring. P-value < 0.05 and absolute fold change > 2 were considered the threshold of differential expression data. Data analyses were performed using R and Bioconductor. RESULTS: We found that 3 differentially expressed miRNAs (miR-10a-5p, miR-10b-5p, miR-629-3p), 6 differentially expressed lncRNAs (TTN-AS1, LINC00641, SNHG14, LINC00894, SNHG1, OIP5-AS1), and 49 differentially expressed mRNAs play a vitally critical role in developing GRE. The expression of GABARAPL1, GRAMD1B, and IQSEC3 were validated more than twofold higher in the GRE group than in the GNE group in the validation cohort. Pathways including ECM receptor interaction and long-term potentiation (LTP) may contribute to the disease's progression. Meanwhile, We built a lncRNA-microRNA-Gene regulatory network with structural and functional significance. CONCLUSION: These findings can offer a fresh perspective on GRE-induced brain network changes.


Asunto(s)
Epilepsia , Glioma , MicroARNs , ARN Largo no Codificante , Redes Reguladoras de Genes , Glioma/complicaciones , Glioma/genética , Glioma/metabolismo , Humanos , Potenciación a Largo Plazo , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Mensajero/genética
3.
Angew Chem Int Ed Engl ; 61(31): e202203546, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35642869

RESUMEN

Recent progress in studying copper-dependent targets and pathways in the context of tumor treatment has provided new insights into therapeutic strategies of leveraging copper-dependent disease vulnerabilities and pharmacological manipulation of intratumor copper transportation to improve chemotherapy. Here, we developed reactive oxygen species (ROS)-sensitive nanoparticles loaded with copper chaperone inhibitor DC_AC50 and cisplatin(IV) prodrug. The released DC_AC50 can promote a remarkable accumulation of intracellular cisplatin and copper through inhibition of the Atox1-ATPase pathways, thereby enhancing the chemotherapeutic effect of cisplatin and inducing significant ROS generation. Excessive ROS then elicits intense endoplasmic reticulin (ER) stress which facilitates the immunogenic cell death (ICD) spurring a sustained immune response. Our study suggests that nanoparticle-mediated copper chaperone inhibition via DC_AC50 can restore the immunogenicity of tumor cells for enhanced chemotherapy and cancer immunotherapy.


Asunto(s)
Nanopartículas , Neoplasias , Línea Celular Tumoral , Cisplatino/metabolismo , Cisplatino/farmacología , Cobre/metabolismo , Chaperonas Moleculares , Neoplasias/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo
4.
Cancer Sci ; 109(7): 2199-2210, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29788550

RESUMEN

Ubiquitin-specific protease 22 (USP22) is a member of the "death-from-cancer" signature, which plays a key role in cancer progression. Previous evidence has shown that USP22 is overexpressed and correlates with poor prognosis in glioma. The effect and mechanism of USP22 in glioma malignancy, especially cancer stemness, remain elusive. Herein, we find USP22 is more enriched in stem-like tumorspheres than differentiated glioma cells. USP22 knockdown inhibits cancer stemness in glioma cell lines. With a cell-penetrating TAT-tag protein, B cell-specific Moloney murine leukemia virus integration site 1 (BMI1), a robust glioma stem-cell marker, is found to mediate the effect of USP22 on glioma stemness. By immunofluorescence, USP22 and BMI1 are found to share similar intranuclear expression in glioma cells. By analysis with immunohistochemistry and bioinformatics, USP22 is found to positively correlate with BMI1 at the post-translational level only rather than at the transcriptional level. By immunoprecipitation and in vivo deubiquitination assay, USP22 is found to interact with and deubiquitinate BMI1 for protein stabilization. Microarray analysis shows that USP22 and BMI1 mutually regulate a series of genes involved in glioma stemness such as POSTN, HEY2, PDGFRA and ATF3. In vivo study with nude mice confirms the role of USP22 in promoting glioma tumorigenesis by regulating BMI1. All these findings indicate USP22 as a novel deubiquitinase of BMI1 in glioma. We propose a working model of the USP22-BMI1 axis, which promotes glioma stemness and tumorigenesis through oncogenic activation. Thus, targeting USP22 might be an effective strategy to treat glioma especially in those with elevated BMI1 expression.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Complejo Represivo Polycomb 1/metabolismo , Tioléster Hidrolasas/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Transformación Celular Neoplásica/genética , Glioma/metabolismo , Xenoinjertos , Humanos , Ratones , Ratones SCID , Proteínas Oncogénicas , Ubiquitina Tiolesterasa
5.
Neurochem Res ; 43(12): 2343-2352, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30311181

RESUMEN

GABA is a dominant inhibitory neurotransmitter in the brain and A type GABA receptor (GABAAR) phosphorylation is critical for GABA-mediated inhibitory effect. However, its role in the neuroprotective effect of sodium valproate (VPA), a prevalent drug for treating patients with epilepsy, remains elusive. The present study was conducted to explore the role of GABAAR phosphorylation in the neuroprotection of VPA against a kainic acid-induced epileptic rat model and the potential molecular mechanisms. Neuronal apoptosis was evaluated by TUNEL assay, PI/Annexin V double staining, caspase-3 activity detection and Bax and Bcl-2 proteins expression via Western blot analysis. The primary rat hippocampal neurons were cultivated and cell viability was measured by CCK8 detection following KA- or free Mg2+-induced neuronal impairment. Our results found that VPA treatment significantly reduced neuronal apoptosis in the KA-induced rat model (including reductions of TUNEL-positive cells, caspase-3 activity and Bax protein expression, and increase of Bcl-2 protein level). In the in vitro experiments, VPA at the concentration of 1 mM for 24 h also increased cell survival and suppressed cell apoptosis in KA- or no Mg2+-induced models via CCK8 assay and PI/Annexin V double staining, respectively. What is more important, the phosphorylation of γ2 subunit at serine 327 residue for GABAAR was found to be robustly enhanced both in the KA-induced epileptic rat model and neuronal cultures following KA exposure after VPA treatment, while no evident alteration was found in terms of GABAAR ß3 phosphorylation (408 or 409 serine residue). Additionally, pharmacological inhibition of protein kinase C (PKC) clearly abrogated the neuroprotective potential of VPA against KA- or free Mg2+-associated neuronal injury, indicating a critical role of PKC in the effect of GABAAR γ2 serine 327 phosphorylation in VPA's protection. In summary, our work reveals that VPA mitigates neuronal apoptosis in KA-triggered epileptic seizures, at least, via augmenting PKC-dependent GABAAR γ2 phosphorylation at serine 327 residue.


Asunto(s)
Epilepsia/metabolismo , Ácido Kaínico/toxicidad , Proteína Quinasa C/metabolismo , Receptores de GABA-A/metabolismo , Serina/metabolismo , Ácido Valproico/uso terapéutico , Animales , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Epilepsia/inducido químicamente , Epilepsia/tratamiento farmacológico , Masculino , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Ratas , Ratas Sprague-Dawley , Ácido Valproico/farmacología
6.
Med Res Rev ; 37(3): 627-661, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27775833

RESUMEN

Glioblastoma (GBM) is regarded as the most common primary intracranial neoplasm. Despite standard treatment with tumor resection and radiochemotherapy, the outcome remains gloomy. It is evident that a combination of oncogenic gain of function and tumor-suppressive loss of function has been attributed to glioma initiation and progression. The ubiquitin-proteasome system is a well-orchestrated system that controls the fate of most proteins by striking a dynamic balance between ubiquitination and deubiquitination of substrates, having a profound influence on the modulation of oncoproteins, tumor suppressors, and cellular signaling pathways. In recent years, deubiquitinating enzymes (DUBs) have emerged as potential anti-cancer targets due to their targeting several key proteins involved in the regulation of tumorigenesis, apoptosis, senescence, and autophagy. This review attempts to summarize recent studies of GBM-associated DUBs, their roles in various cellular processes, and discuss the relation between DUBs deregulation and gliomagenesis, especially how DUBs regulate glioma stem cells pluripotency, microenvironment, and resistance of radiation and chemotherapy through core stem-cell transcriptional factors. We also review recent achievements and progress in the development of potent and selective reversible inhibitors of DUBs, and attempted to find a potential GBM treatment by DUBs intervention.


Asunto(s)
Enzimas Desubicuitinizantes/metabolismo , Glioblastoma/enzimología , Glioblastoma/terapia , Terapia Molecular Dirigida , Animales , Carcinogénesis/patología , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Humanos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología
7.
Tumour Biol ; 37(11): 14403-14413, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27629290

RESUMEN

With the development of whole genome and transcriptome sequencing technologies, a growing body of long non-coding RNAs (lncRNAs) has been identified and is receiving increasing attention. LncRNAs are non-protein encoding transcripts whose functions are crucial for advancing our comprehensive understanding of biological processes in human health and diseases, specifically glioma. It has been established that lncRNAs are differently expressed in the central nervous system and may play a vital role in glioma. As of June 2016, 20 lncRNAs have been identified that may play a role in glioma pathogenesis. Investigation into the role of lncRNAs in glioma may help to identify potential biomarkers which can improve the diagnosis and treatment of glioma. In this paper, we review current understanding of the function of lncRNAs in glioma initiation and progression.


Asunto(s)
Sistema Nervioso Central/patología , Glioma/genética , Glioma/patología , ARN Largo no Codificante/genética , Biomarcadores de Tumor/genética , Progresión de la Enfermedad , Humanos
8.
Cell Mol Neurobiol ; 36(6): 915-925, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26440805

RESUMEN

Oxidative glutamate toxicity is involved in diverse neurological disorders including epilepsy and ischemic stroke. Our present work aimed to assess protective effects of huperzine A (HupA) against oxidative glutamate toxicity in a mouse-derived hippocampal HT22 cells and explore its potential mechanisms. Cell survival and cell injury were analyzed by MTT method and LDH release assay, respectively. The production of ROS was measured by detection kits. Protein expressions of BDNF, phosphor-TrkB (p-TrkB), TrkB, phosphor-Akt (p-Akt), Akt, phosphor-mTOR (p-mTOR), mTOR, phosphor-p70s6 (p-p70s6) kinase, p70s6 kinase, Bcl-2, Bax, and ß-actin were assayed via Western blot analysis. Enzyme-linked immunosorbent assay was employed to measure the contents of nerve growth factor, brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and neurotrophin-4 (NT-4). Our findings illustrated 10 µM HupA for 24 h significantly protected HT22 from cellular damage and suppressed the generation of ROS. Additionally, after treating with LY294002 or wortmannin [the selective inhibitors of phosphatidylinositol 3 kinase (PI3K)], HupA dramatically prevented the down-regulations of p-Akt, p-mTOR, and p-p70s6 kinase in HT22 cells under oxidative toxicity. Furthermore, it was observed that the protein levels of BDNF and p-TrkB were evidently enhanced after co-treatment with HupA and glutamate in HT22 cells. The elevations of p-Akt and p-mTOR were abrogated under toxic conditions after blockade of TrkB by TrkB IgG. Cellular apoptosis was significantly suppressed (decreased caspase-3 activity and enhanced Bcl-2 protein level) after HupA treatment. It was concluded that HupA attenuated oxidative glutamate toxicity in murine hippocampal HT22 cells via activating BDNF/TrkB-dependent PI3K/Akt/mTOR signaling pathway.


Asunto(s)
Alcaloides/farmacología , Hipocampo/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Sesquiterpenos/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ácido Glutámico/farmacología , Hipocampo/metabolismo , Ratones , Fosfatidilinositol 3-Quinasa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor trkB/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
9.
Int J Mol Sci ; 17(11)2016 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-27801851

RESUMEN

Current treatment methods for patients diagnosed with gliomas have shown limited success. This is partly due to the lack of prognostic genes available to accurately predict disease outcomes. The aim of this study was to investigate novel prognostic genes based on the molecular profile of tumor samples and their correlation with clinical parameters. In the current study, microarray data (GSE4412 and GSE7696) downloaded from Gene Expression Omnibus were used to identify differentially expressed prognostic genes (DEPGs) by significant analysis of microarray (SAM) between long-term survivors (>2 years) and short-term survivors (≤2 years). DEPGs generated from these two datasets were intersected to obtain a list of common DEPGs. The expression of a subset of common DEPGs was then independently validated by real-time reverse transcription quantitative PCR (qPCR). Survival value of the common DEPGs was validated using known survival data from the GSE4412 and TCGA dataset. After intersecting DEPGs generated from the above two datasets, three genes were identified which may potentially be used to determine glioma patient prognosis. Independent validation with glioma patients tissue (n = 70) and normal brain tissue (n = 19) found PPIC, EMP3 and CHI3L1 were up-regulated in glioma tissue. Survival value validation showed that the three genes correlated with patient survival by Kaplan-Meir analysis, including grades, age and therapy.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Proteína 1 Similar a Quitinasa-3/biosíntesis , Ciclofilina C/biosíntesis , Glioma/genética , Glicoproteínas de Membrana/biosíntesis , Adulto , Factores de Edad , Anciano , Biomarcadores de Tumor/genética , Proteína 1 Similar a Quitinasa-3/genética , Ciclofilina C/genética , Dacarbazina/administración & dosificación , Dacarbazina/análogos & derivados , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Glioma/tratamiento farmacológico , Glioma/patología , Glioma/radioterapia , Humanos , Estimación de Kaplan-Meier , Masculino , Glicoproteínas de Membrana/genética , Análisis por Micromatrices , Persona de Mediana Edad , Clasificación del Tumor , Pronóstico , Temozolomida
10.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 40(9): 955-9, 2015 Sep.
Artículo en Zh | MEDLINE | ID: mdl-26408613

RESUMEN

OBJECTIVE: To investigate the neuroprotective effects of osthole (OST) on glutamate-induced toxicity in hippocampal HT22 cells and to explore the correlation between the protection and phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt) signaling pathway.
 METHODS: The cell injury model of HT22 was induced by glutamate and the cell viability was detected by MTS assay. The lactate dehydrogenase (LDH) release and the caspase-3 activity were determined by commercial kits. Western blot analysis was utilized to detect the protein levels of PI3K, Akt, p-PI3K and p-Akt. 
 RESULTS: OST markedly improved the cell survival and decreased the LDH release in glutamate-treated HT22 cells in a dose-dependent manner. Furthermore, the levels of p-PI3K and p-Akt proteins were significantly increased in glutamate and OST-co-treated HT22 cells. The effect of OST on p-Akt phosphorylation in HT22 cells was attenuated in the presence of PI3K specific inhibitor (LY294002).
 CONCLUSION: OST protects HT22 cells from glutamate excitotoxicity through a mechanism involving the activation of PI3K/Akt signaling pathway.


Asunto(s)
Cumarinas/farmacología , Ácido Glutámico/efectos adversos , Fármacos Neuroprotectores/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Animales , Caspasa 3/metabolismo , Línea Celular , Supervivencia Celular , Cromonas/farmacología , Hipocampo/citología , Ratones , Morfolinas/farmacología , Fosforilación
11.
Neurol Sci ; 35(8): 1261-8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24590842

RESUMEN

Baicalein has been shown to possess various pharmacological actions. The current work was designed to assess the neuroprotection of baicalein against cognitive deficits in epilepsy-like tremor rat (TRM). Epileptic characteristics and memory functions were assessed by electroencephalograms recording and Morris water maze test, respectively. The changes of oxidative indicators including malondialdehyde (MDA), catalase (CAT), Cu/Zn-superoxide dismutase (Cu/Zn-SOD), Mn-SOD, glutathione (GSH), glutathione peroxidase (GSH-PX) and 8-isoprostane were measured using corresponding commercial kits. Real-time RT-PCR and immunoassay were employed to detect activities of various inflammatory mediators such as NF-κB p65, TNF-α, IL-1ß, IL-6 and IL-10. Western blot analysis was performed to determine heat shock protein (HSP) 70 and mitogen-activated protein kinases (MAPKs) (including ERK, JNK and p38) proteins. Our results illustrated that baicalein significantly ameliorated epileptiform activity and cognitive deficits in TRM. Besides, reduced oxidative stress and inflammatory responses were also found in TRM treated with baicalein. Furthermore, there were evident alterations of HSP70 and MAPK cascades at protein levels after 14-day pretreatment with baicalein. It was concluded that the neuroprotective effect of baicalein against cognitive dysfunction might be associated with suppressing oxidative stress, inhibiting inflammation and mediating HSP70 as well as MAPK cascades in absence-like TRM.


Asunto(s)
Química Encefálica/efectos de los fármacos , Medicamentos Herbarios Chinos/uso terapéutico , Epilepsia Tipo Ausencia/psicología , Flavanonas/uso terapéutico , Discapacidades para el Aprendizaje/prevención & control , Fármacos Neuroprotectores/uso terapéutico , Nootrópicos/uso terapéutico , Convulsiones/psicología , Animales , Antiinflamatorios/uso terapéutico , Antioxidantes/uso terapéutico , Citocinas/análisis , Evaluación Preclínica de Medicamentos , Electroencefalografía , Epilepsia Tipo Ausencia/genética , Epilepsia Tipo Ausencia/metabolismo , Proteínas de Choque Térmico/análisis , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Mediadores de Inflamación/análisis , Discapacidades para el Aprendizaje/tratamiento farmacológico , Discapacidades para el Aprendizaje/etiología , Discapacidades para el Aprendizaje/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/análisis , Proteínas del Tejido Nervioso/análisis , Estrés Oxidativo/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Mutantes , Convulsiones/genética , Convulsiones/metabolismo , Transducción de Señal/efectos de los fármacos
12.
Int J Mol Sci ; 15(5): 7667-83, 2014 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-24857910

RESUMEN

The present study was designed to probe the effects of Huperzine A (HupA) on diabetes-associated cognitive decline (DACD) using a streptozotocin (STZ)-injected rat model. Diabetic rats were treated with HupA (0.05 and 0.1 mg/kg) for seven weeks. Memory functions were evaluated by the water maze test. Nissl staining was selected for detecting neuronal loss. Protein and mRNA levels of brain-derived neurotrophic factor (BDNF) were analyzed by ELISA and real-time PCR, respectively. The activities of choline acetylase (ChAT), Acetylcholinesterase (AChE), malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), catalase (CAT), NF-κB p65 unit, TNF-α, IL-1ß, IL-6 and caspase-3 were measured using corresponding kits. After seven weeks, diabetic rats exhibited remarkable reductions in: body weight, percentage of time spent in target quadrant, number of times crossing the platform, ChAT and BDNF levels, SOD, GSH-Px and CAT accompanied with increases in neuronal damage, plasma glucose levels, escape latency, mean path length, AChE, MDA level as well as CAT, NF-κB p65 unit, TNF-α, IL-1ß, IL-6 and caspase-3 in cerebral cortex and hippocampus. Supplementation with HupA significantly and dose-dependently reversed the corresponding values in diabetes. It is concluded that HupA ameliorates DACD via modulating BDNF, oxidative stress, inflammation and apoptosis.


Asunto(s)
Alcaloides/farmacología , Memoria/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Sesquiterpenos/farmacología , Alcaloides/química , Alcaloides/uso terapéutico , Animales , Glucemia/análisis , Peso Corporal/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/análisis , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Caspasa 3/metabolismo , Trastornos del Conocimiento/complicaciones , Trastornos del Conocimiento/tratamiento farmacológico , Citocinas/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Ensayo de Inmunoadsorción Enzimática , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Oxidorreductasas/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Sesquiterpenos/química , Sesquiterpenos/uso terapéutico , Estreptozocina/toxicidad
13.
Int J Mol Sci ; 15(12): 23408-17, 2014 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-25522171

RESUMEN

Previous studies reported that the proline-rich transmembrane protein 2 (PRRT2) gene was identified to be related to paroxysmal kinesigenic dyskinesia (PKD), infantile convulsions with PKD, PKD with migraine and benign familial infantile epilepsy (BFIE). The present study explores whether the PRRT2 mutation is a potential cause of febrile seizures, including febrile seizures plus (FS+), generalized epilepsy with febrile seizures plus (GEFS+) and Dravet syndrome (DS); thus, it may provide a new drug target for personalized medicine for febrile seizure patients. We screened PRRT2 exons in a cohort of 136 epileptic patients with febrile seizures, including FS+, GEFS+ and DS. PRRT2 genetic mutations were identified in 25 out of 136 (18.4%) febrile seizures in epileptic patients. Five loss-of-function and coding missense mutations were identified: c.649delC (p.R217Efs*12), c.649_650insC (p.R217Pfs*8), c.412C>G (p.Pro138Ala), c.439G>C (p.Asp147His) and c.623C>A (p.Ser208Tyr). PRRT2 variants were probably involved in the etiology of febrile seizures in epileptic patients.


Asunto(s)
Estudios de Asociación Genética , Proteínas de la Membrana/genética , Mutación , Proteínas del Tejido Nervioso/genética , Convulsiones Febriles/genética , Adolescente , Adulto , Edad de Inicio , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Convulsiones Febriles/diagnóstico , Adulto Joven
14.
Adv Mater ; 36(7): e2306419, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37796042

RESUMEN

Promoting innate immunity through pyroptosis induction or the cyclic GMP-AMP synthase-stimulator of interferon gene (cGAS-STING) pathway activation has emerged as a potent approach to counteract the immunosuppressive tumor microenvironment and elicit systemic antitumor immunity. However, current pyroptosis inducers and STING agonists often suffer from limitations including instability, unpredictable side effects, or inadequate intracellular expression of gasdermin and STING. Here, a tumor-specific nanotheranostic platform that combines photodynamic therapy (PDT) with epigenetic therapy to simultaneously activate pyroptosis and the cGAS-STING pathway in a light-controlled manner is constructed. This approach involves the development of oxidation-sensitive nanoparticles (NP1) loaded with the photosensitizer TBE, along with decitabine nanomicelles (NP2). NP2 enables the restoration of STING and gasdermin E (GSDME) expression, while NP1-mediated PDT facilitates the release of DNA fragments from damaged mitochondria to potentiate the cGAS-STING pathway, and promotes the activation of caspase-3 to cleave the upregulated GSDME into pore-forming GSDME-N terminal. Subsequently, the released inflammatory cytokines facilitate the maturation of antigen-presentation cells, triggering T cell-mediated antitumor immunity. Overall, this study presents an elaborate strategy for simultaneous photoactivation of pyroptosis and the cGAS-STING pathway, enabling targeted photoimmunotherapy in immunotolerant tumors. This innovative approach holds significant promise in overcoming the limitations associated with existing therapeutic modalities and represents a valuable avenue for future clinical applications.


Asunto(s)
Interferones , Neoplasias , Humanos , Gasderminas , Piroptosis , Nanomedicina Teranóstica , Neoplasias/tratamiento farmacológico , Epigénesis Genética , Nucleotidiltransferasas , Microambiente Tumoral
15.
J Adv Res ; 63: 73-90, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39048074

RESUMEN

INTRODUCTION: Our previous work reveals a critical role of activation of neuronal Alox5 in exacerbating brain injury post seizures. However, whether neuronal Alox5 impacts the pathological process of epilepsy remains unknown. OBJECTIVES: To prove the feasibility of neuron-specific deletion of Alox5 via CRISPR-Cas9 in the blockade of seizure onset and epileptic progression. METHODS: Here, we employed a Clustered regularly interspaced short-palindromic repeat-associated proteins 9 system (CRISPR/Cas9) system delivered by adeno-associated virus (AAV) to specifically delete neuronal Alox5 gene in the hippocampus to explore its therapeutic potential in various epilepsy mouse models and possible mechanisms. RESULTS: Neuronal depletion of Alox5 was successfully achieved in the brain. AAV delivery of single guide RNA of Alox5 in hippocampus resulted in reducing seizure severity, delaying epileptic progression and improving epilepsy-associated neuropsychiatric comorbidities especially anxiety, cognitive deficit and autistic-like behaviors in pilocarpine- and kainic acid-induced temporal lobe epilepsy (TLE) models. In addition, neuronal Alox5 deletion also reversed neuron loss, neurodegeneration, astrogliosis and mossy fiber sprouting in TLE model. Moreover, a battery of tests including analysis of routine blood test, hepatic function, renal function, routine urine test and inflammatory factors demonstrated no noticeable toxic effect, suggesting that Alox5 deletion possesses the satisfactory biosafety. Mechanistically, the anti-epileptic effect of Alox5 deletion might be associated with reduction of glutamate level to restore excitatory/inhibitory balance by reducing CAMKII-mediated phosphorylation of Syn ISer603. CONCLUSION: Our findings showed the translational potential of AAV-mediated delivery of CRISPR-Cas9 system including neuronal Alox5 gene for an alternative promising therapeutic approach to treat epilepsy.


Asunto(s)
Araquidonato 5-Lipooxigenasa , Sistemas CRISPR-Cas , Modelos Animales de Enfermedad , Epilepsia , Hipocampo , Neuronas , Animales , Ratones , Araquidonato 5-Lipooxigenasa/metabolismo , Araquidonato 5-Lipooxigenasa/genética , Neuronas/metabolismo , Hipocampo/metabolismo , Epilepsia/genética , Masculino , Ratones Endogámicos C57BL , Eliminación de Gen , Epilepsia del Lóbulo Temporal/genética , Dependovirus/genética , Pilocarpina
16.
CNS Neurosci Ther ; 30(8): e14911, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39145422

RESUMEN

BACKGROUND: Epilepsy is a widespread central nervous system disorder with an estimated 50 million people affected globally. It is characterized by a bimodal incidence peak among infants and the elderly and is influenced by a variety of risk factors, including a significant genetic component. Despite the use of anti-epileptic drugs (AEDs), drug-refractory epilepsy develops in about one-third of patients, highlighting the need for alternative therapeutic approaches. AIMS: The primary aim of this study was to evaluate the neuroprotective effects of troglitazone (TGZ) in epilepsy and to explore the potential mechanisms underlying its action. METHODS: We employed both in vitro and in vivo models to assess TGZ's effects. The in vitro model involved glutamate-induced toxicity in HT22 mouse hippocampal neurons, while the in vivo model used kainic acid (KA) to induce epilepsy in mice. A range of methods, including Hoechst/PI staining, CCK-8 assay, flow cytometry, RT-PCR analysis, Nissl staining, scanning electron microscopy, and RNA sequencing, were utilized to assess various parameters such as cellular damage, viability, lipid-ROS levels, mitochondrial membrane potential, mRNA expression, seizure grade, and mitochondrial morphology. RESULTS: Our results indicate that TGZ, at doses of 5 or 20 mg/kg/day, significantly reduces KA-induced seizures and neuronal damage in mice by inhibiting the process of ferroptosis. Furthermore, TGZ was found to prevent changes in mitochondrial morphology. In the glutamate-induced HT22 cell damage model, 2.5 µM TGZ effectively suppressed neuronal ferroptosis, as shown by a reduction in lipid-ROS accumulation, a decrease in mitochondrial membrane potential, and an increase in PTGS2 expression. The anti-ferroptotic effect of TGZ was confirmed in an erastin-induced HT22 cell damage model as well. Additionally, TGZ reversed the upregulation of Plaur expression in HT22 cells treated with glutamate or erastin. The downregulation of Plaur expression was found to alleviate seizures and reduce neuronal damage in the mouse hippocampus. CONCLUSION: This study demonstrates that troglitazone has significant therapeutic potential in the treatment of epilepsy by reducing epileptic seizures and the associated brain damage through the inhibition of neuronal ferroptosis. The downregulation of Plaur expression plays a crucial role in TGZ's anti-ferroptotic effect, offering a promising avenue for the development of new epilepsy treatments.


Asunto(s)
Epilepsia , Ferroptosis , Fármacos Neuroprotectores , Troglitazona , Animales , Ratones , Epilepsia/tratamiento farmacológico , Epilepsia/inducido químicamente , Ferroptosis/efectos de los fármacos , Ferroptosis/fisiología , Fármacos Neuroprotectores/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/patología , Hipocampo/metabolismo , Ácido Glutámico/metabolismo , Masculino , Ácido Kaínico/toxicidad , Ratones Endogámicos C57BL , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico
17.
Biomedicines ; 12(1)2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38275418

RESUMEN

Circular RNA (circRNA) plays a very important regulatory role in a variety of human malignancies such as non-small-cell lung cancer (NSCLC). In the current study, we explored the role of hsa_circ_0092856 in the progression of NSCLC. We screened CircRNA from the eIF3a gene in the Circbase database. The biological functions of hsa_circ_0092856 in NSCLC were analyzed via qRT-PCR, a CCK-8 assay, a plate cloning experiment, scratch testing, a transwell chamber experiment, an RNA nuclear mass separation experiment, an RIP experiment, and a Western blot test. The results showed that hsa_circ_0092856 was highly expressed in NSCLC cells, and the knockdown of hsa_circ_0092856 could inhibit the proliferation, migration, and invasion of NSCLC cells. The overexpression of hsa_circ_0092856 has the opposite effect. The expression of eIF3a also changed with the change in hsa_circ_0092856. These results suggest that hsa_circ_0092856 may play a key role in the progression of NSCLC by regulating the expression of eIF3a.

18.
Brain Res Bull ; 210: 110928, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38493836

RESUMEN

Epilepsy-associated cognitive disorder (ECD), a prevalent comorbidity in epilepsy patients, has so far uncharacterized etiological origins. Our prior work revealed that lysyl oxidase (Lox) acted as a novel contributor of ferroptosis, a recently discovered cell death mode in the regulation of brain function. However, the role of Lox-mediated ferroptosis in ECD remains unknown. ECD mouse model was established 2 months later following a single injection of kainic acid (KA) for. After chronic treatment with KA, mice were treated with different doses (30 mg/kg, 100 mg/kg and 300 mg/kg) of Lox inhibitor BAPN. Additionally, hippocampal-specific Lox knockout mice was also constructed and employed to validate the role of Lox in ECD. Cognitive functions were assessed using novel object recognition test (NOR) and Morris water maze test (MWM). Protein expression of phosphorylated cAMP-response element binding (CREB), a well-known molecular marker for evaluation of cognitive performance, was also detected by Western blot. The protein distribution of Lox was analyzed by immunofluorescence. In KA-induced ECD mouse model, ferroptosis process was activated according to upregulation of 4-HNE protein and a previously discovered ferroptosis in our group, namely, Lox was remarkably increased. Pharmacological inhibition of Lox by BAPN at the dose of 100 mg/kg significantly increased the discrimination index following NOR test and decreased escape latency as well as augmented passing times within 60 s following MWM test in ECD mouse model. Additionally, deficiency of Lox in hippocampus also led to pronounced improvement of deficits in ECD model. These findings indicate that the ferroptosis regulatory factor, Lox, is activated in ECD. Ablation of Lox by either pharmacological intervention or genetic manipulation ameliorates the impairment in ECD mouse model, which suggest that Lox serves as a promising therapeutic target for treating ECD in clinic.


Asunto(s)
Disfunción Cognitiva , Epilepsia , Humanos , Ratones , Animales , Proteína-Lisina 6-Oxidasa/genética , Proteína-Lisina 6-Oxidasa/metabolismo , Aminopropionitrilo/farmacología , Regulación de la Expresión Génica , Modelos Animales de Enfermedad , Disfunción Cognitiva/tratamiento farmacológico
19.
Eur J Surg Oncol ; 49(9): 106902, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37076410

RESUMEN

Despite the wide reportage of prognostic factors for glioblastoma (GBM), it is difficult to determine how these factors interact to affect patients' survival. To determine the combination of prognostic factors, we retrospectively analyzed the clinic data of 248 IDH wild-type GBM patients and built a novel prediction model. The survival variables of patients were identified via univariate and multivariate analyses. In addition, the score prediction models were constructed by combining classification and regression tree (CART) analysis with Cox regression analysis. Finally, the prediction model was internally validated using the bootstrap method. Patients were followed for a median of 34.4 (interquartile range, 26.1-46.0) months. Multivariate analysis identified gross total resection (GTR) (HR 0.50, 95% CI: 0.38-0.67), unopened ventricles (HR 0.75 [0.57-0.99]), and MGMT methylation (HR 0.56 [0.41-0.76]) as favorable independent prognostic factors for PFS. GTR (HR 0.67 [0.49-0.92]), unopened ventricles (HR 0.60 [0.44-0.82]), and MGMT methylation (HR 0.54 [0.38-0.76]) were favorable independent prognostic factors for OS. In the process of building the model, we incorporated GTR, ventricular opening, MGMT methylation status, and age. The model had six and five terminal nodules in PFS and OS respectively. We grouped terminal nodes with similar hazard ratios together to form three sub-groups with different PFS and OS (P < 0.001). After the internal verification of bootstrap method, the model had a good fitting and calibration. GTR, unopened ventricles, and MGMT methylation were independently associated with more satisfactory survival. The novel score prediction model which we construct can provide a prognostic reference for GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/genética , Glioblastoma/cirugía , Pronóstico , Estudios Retrospectivos , Metilasas de Modificación del ADN/genética , Metilasas de Modificación del ADN/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Neoplasias Encefálicas/cirugía , Metilación de ADN
20.
Genes Dis ; 10(3): 771-785, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37396555

RESUMEN

Vascular remodeling and angiogenesis are two key processes in the maintenance of vascular homeostasis and involved in a wide array of vascular pathologies. Following these processes, extracellular matrix (ECM) provides the mechanical foundation for vascular walls. Lysyl oxidase (LOX), the key matrix-modifying enzyme, has been demonstrated to significantly affect structural abnormality and dysfunction in the blood vessels. The role of LOX in vascular remodeling and angiogenesis has always been the subject in the current medical research. Therefore, we presently make a summarization of the biosynthesis of LOX and the mechanisms involved in vascular remodeling and angiogenesis, as well as the role of LOX in diseases associated with vascular abnormalities and the therapeutic potential via targeting LOX. In particular, we give a proposal that LOX likely reshapes matrisome-associated genes expressions in the regulation of vascular remodeling and angiogenesis, which serves as a mechanistic insight into the critical role of LOX in these two aspects. Additionally, LOX has also dual effects on the vascular dysfunction, namely, inhibition of LOX for improving hypertension, restenosis and malignant tumor while activation of LOX for curing arterial aneurysm and dissection. LOX-targeted therapy may provide a promising therapeutic strategy for the treatment of various vascular pathologies associated with vascular remodeling and angiogenesis.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda