Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Pathol ; 193(11): 1845-1862, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37517685

RESUMEN

The transcription factor forkhead box protein (FOX)-O3 is a core regulator of cellular homeostasis, stress response, and longevity. The cellular localization of FOXO3 is closely related to its function. Herein, the role of FOXO3 in cataract formation was explored. FOXO3 showed nuclear translocation in lens epithelial cells (LECs) arranged in a single layer on lens capsule tissues from both human cataract and N-methyl-N-nitrosourea (MNU)-induced rat cataract, also in MNU-injured human (H)-LEC lines. FOXO3 knockdown inhibited the MNU-induced increase in expression of genes related to cell cycle arrest (GADD45A and CCNG2) and apoptosis (BAK and TP53). H2 is highly effective in reducing oxidative impairments in nuclear DNA and mitochondria. When H2 was applied to MNU-injured HLECs, FOXO3 underwent cleavage by MAPK1 and translocated into mitochondria, thereby increasing the transcription of oxidative phosphorylation-related genes (MTCO1, MTCO2, MTND1, and MTND6) in HLECs. Furthermore, H2 mediated the translocation of FOXO3 from the nucleus to the mitochondria within the LECs of cataract capsule tissues of rats exposed to MNU. This intervention ameliorated MNU-induced cataracts in the rat model. In conclusion, there was a correlation between the localization of FOXO3 and its function in cataract formation. It was also determined that H2 protects HLECs from injury by leading FOXO3 mitochondrial translocation via MAPK1 activation. Mitochondrial FOXO3 can increase mtDNA transcription and stabilize mitochondrial function in HLECs.

2.
Eye (Lond) ; 37(12): 2488-2493, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36543942

RESUMEN

AIMS: To detect mutations in juvenile-onset open-angle glaucoma in a Chinese family and to describe the characteristic ophthalmic phenotypes of this pedigree. METHODS: There were 14 individuals in this four-generation pedigree. All living members of the family underwent comprehensive ophthalmic examinations. Five patients presented with elevated intraocular pressures. All of them shared early-onset disease, with a mean onset age of 14.4 years and continuing aggressive damage to their optic nerves. Hyperpigmentation in the trabecular meshwork and sometimes-broad iris processes were noted in this family using gonioscopy. All exons of candidate genes (MYOC, OPTN, CYP1B1) were amplified using the polymerase chain reaction, and analysed with an ABI 3700XL Genetic Analyser. RESULTS: A heterozygous missense mutation in exon 3 (c.733 T > G) of the MYOC gene was found in the five JOAG patients and one 7-year-old boy with no ophthalmic manifestation of glaucoma, but it was absent in other members of the family and in the controls. This mutation resulted in a transversion of cysteine to glycine (Cys245Gly). CONCLUSIONS: We concluded the novel MYOC c.733 T > G mutation found in a Chinese family with JOAG caused a severe type of JOAG exhibiting early onset, high IOP, and severe optic nerve damage. Interestingly, unlike other reported MYOC mutation families, our patients exhibited marked angle pigmentation and iris processes.


Asunto(s)
Proteínas del Ojo , Glaucoma de Ángulo Abierto , Humanos , Pueblos del Este de Asia , Ojo , Proteínas del Ojo/genética , Glaucoma de Ángulo Abierto/genética , Glaucoma de Ángulo Abierto/diagnóstico , Mutación , Linaje
3.
J Thorac Oncol ; 17(10): 1216-1226, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35798241

RESUMEN

INTRODUCTION: As a novel third-generation EGFR tyrosine kinase inhibitor (TKI), SH-1028 (formerly oritinib) is developed to inhibit both sensitizing EGFR mutations and EGFR T790M mutation. METHODS: This was a multicenter, single-arm, open-label, phase 2 trial (NCT03823807). Eligible patients were those with advanced NSCLC with centrally confirmed EGFR T790M mutation who progressed after first- or second-generation EGFR TKIs or with primary EGFR T790M mutations. Each patient received SH-1028 tablets orally at 200 mg/d until disease progression or intolerable toxicity. Tumor response was evaluated every 6 weeks per the Response Evaluation Criteria in Solid Tumors, version 1.1. The primary end point was objective response rate by an independent review committee. The secondary end points were progression-free survival, overall survival (OS), disease control rate, safety, and so on. RESULTS: A total of 286 patients with EGFR T790M-positive advanced NSCLC were enrolled in this study, including 59 patients in part A (dose-verification study) and 227 patients in part B (second-line registration study). By data cutoff on September 17, 2021, the independent review committee-assessed objective response rate was 55.9% (95% confidence interval [CI]: 42.4-68.8) in part A and 60.4% (95% CI: 53.7-66.8) in part B. The median progression-free survival was 12.4 months (95% CI: 8.3-20.8) in part A and 12.6 months (95% CI: 9.7-15.3) in part B. The median OS was 26.0 months (95% CI: 23.3-not reached) in part A, and OS was immature in part B. Among the 286 patients, 44 of them experienced at least one grade 3 or higher treatment-related adverse event, with the most common ones as increased serum creatinine phosphokinase level (13 [4.5%]), diarrhea (six [2.1%]), and prolonged QT interval (three [1.0%]). Treatment-related skin rash was reported in 26 patients (9.1%), all grade 1 or 2. There was no interstitial lung disease reported in this study. CONCLUSIONS: SH-1028 is efficacious and tolerable in second-line treatment of patients with advanced NSCLC with positive EGFR T790M.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Compuestos de Anilina/efectos adversos , Carcinoma de Pulmón de Células no Pequeñas/inducido químicamente , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Creatinina , Receptores ErbB/genética , Humanos , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Mutación , Inhibidores de Proteínas Quinasas/uso terapéutico
4.
Front Physiol ; 13: 995353, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36589420

RESUMEN

Purpose: The aim of the study was to investigate microcirculation changes in the macula evaluated by optical coherence tomography angiography (OCTA)in patients receiving anatomical repair after surgery for rhegmatogenous retinal detachment (RRD). Methods: A literature search was conducted in PubMed, EMBASE, Web of Science and the Cochrane Library. Studies including patients with macula-on or macula-off RRD and repaired successfully through primary surgery were selected. Foveal avascular zone (FAZ) area and macular vascular density (VD) in both the superficial capillary plexus (SCP) and deep capillary plexus (DCP) were analyzed using RevMan 5.4 software. Results: Twelve studies including 430 RRD eyes and 430 control eyes were selected. In eyes with macula-on RRD, FAZ area, VD in the foveal SCP and DCP, and VD in the parafoveal SCP and DCP were not altered compared with control eyes, after the retina was reattached. In eyes with macula-off RRD that was repaired successfully through surgery, FAZ area in the DCP (0.13 mm2, 95% CI: 0.02 to 0.25, p = 0.02) remained enlarged compared with control eyes. Meanwhile, VD in the foveal DCP was also significantly reduced (-3.12%, 95% CI: -6.15 to -0.09%, p = 0.04), even though retinal reattachment was achieved by surgery in eyes with macula-off RRD. Conclusion: In patients with macula-off rhegmatogenous retinal detachment, foveal avascular zone area in the deep capillary plexuses was enlarged and vascular density in the foveal deep capillary plexus was reduced, even after the retina was successfully reattached through a primary surgery.

5.
Invest Ophthalmol Vis Sci ; 62(9): 2, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34196654

RESUMEN

Purpose: Previous work by our group has demonstrated the value of N-methyl-N-nitrosourea (MNU)-induced corneal endothelial decompensation in animal models. The aim of this study was to investigate the effect of molecular hydrogen (H2) on MNU-induced corneal endothelial cell (CEC) injury and the underlying mechanism. Methods: MNU-induced animal models of CEC injury were washed with hydrogen-rich saline (HRS) for 14 days. Immunofluorescence staining, immunohistochemical staining, and corneal endothelial assessment were applied to determine architectural and cellular changes on the corneal endothelium following HRS treatment. MNU-induced cell models of CEC injury were co-cultured with H2. The effect of H2 was examined using morphological and functional assays. Results: It was shown that MNU could inhibit the proliferation and specific physiological functions of CECs by increasing apoptosis and decreasing the expression of ZO-1 and Na+/K+-ATPase, whereas H2 improved the proliferation and physiological function of CECs by anti-apoptosis. Cell experiments further confirmed that H2 could reverse MNU damage to CECs by decreasing oxidative stress injury, interfering with the NF-κB/NLRP3 pathway and the FOXO3a/p53/p21 pathway. Conclusions: This study suggests that topical application of H2 could protect CECs against corneal damage factors through anti-apoptotic effect, reduce the incidence and severity of corneal endothelial decompensation, and maintain corneal transparency.


Asunto(s)
Apoptosis/efectos de los fármacos , Lesiones de la Cornea/inducido químicamente , Endotelio Corneal/metabolismo , Hidrógeno/farmacología , Estrés Oxidativo , Regulación hacia Arriba , Animales , Recuento de Células , Células Cultivadas , Lesiones de la Cornea/metabolismo , Lesiones de la Cornea/patología , Modelos Animales de Enfermedad , Endotelio Corneal/efectos de los fármacos , Endotelio Corneal/patología , Masculino , Metilnitrosourea/toxicidad , Conejos , Ratas , Activación Transcripcional
6.
Oncol Lett ; 21(3): 216, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33688375

RESUMEN

[This corrects the article DOI: 10.3892/ol.2015.3719.].

7.
Invest Ophthalmol Vis Sci ; 61(8): 38, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32721019

RESUMEN

Purpose: N-methyl-N-nitrosourea (MNU) is an alkylating toxicant with potent mutagenic ability. This study was designed to induce apoptosis in lens epithelial cells (LECs) and corneal endothelial cells (CECs) via MNU administration. We sought to build ocular disease models of cataract and corneal endothelial decompensation. Methods: MNU was delivered into the intraperitoneal cavities of neonatal rats and the anterior chambers of adult rabbits. The MNU-treated animals were then subjected to a series of functional and morphological analyses at various time points. Results: MNU treatment induced pervasive apoptosis of LECs and CECs. These effects were dose and time dependent. Mature cataracts were found in neonatal rats 3 weeks after MNU treatment. Histological analysis revealed that MNU toxicity induced swelling, vacuolation, and liquefaction in lens fibers of MNU-treated rats. Pentacam examination showed that the average density of rat lens increased significantly after MNU administration. Terminal deoxynucleotidyl transferase-mediated nick end labeling (TUNEL) analysis showed pervasive apoptotic staining in the lenses of MNU-treated rats. In rabbit eyes, intracameral treatment with MNU induced corneal edema and significantly increased central corneal thickness, which peaked at P14. Morphological and immunohistochemical analysis showed that CECs were effectively ablated in the MNU-treated rabbits. The expression of 8-OHdG increased significantly in the cornea of MNU-treated rabbits, compared with vehicle-treated controls. Conclusions: MNU is sufficient to induce ocular cell apoptosis in animal models. These models of MNU-induced cataract and corneal endothelial decompensation represent valuable tools for efforts to develop relevant therapies.


Asunto(s)
Enfermedades de la Córnea , Modelos Animales de Enfermedad , Enfermedades del Cristalino , Metilnitrosourea/farmacología , Conejos , Ratas , Alquilantes/farmacología , Animales , Apoptosis/efectos de los fármacos , Enfermedades de la Córnea/etiología , Enfermedades de la Córnea/patología , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Enfermedades del Cristalino/etiología , Enfermedades del Cristalino/patología , Reproducibilidad de los Resultados
8.
Mol Med Rep ; 21(1): 240-248, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31746431

RESUMEN

Ocular albinism type 1 (OA1) is a genetic disorder characterized by reduced eye pigmentation and nystagmus, which is often accompanied by decreased visual acuity, strabismus and other symptoms, whereas skin and hair color remain normal. The present study aimed to assess the clinical features and perform genotype analysis of a family with OA1, and to determine the disease­causing mutation. A total of 18 family members (nine affected patients and nine normal subjects) from Hainan, China, were recruited to the present study in December 2017. A detailed clinical ophthalmic examination was performed for all participants, including a visual acuity test, anterior segment slit lamp examination, eye fundus examination and optical coherence tomography. Mutations in the G protein­coupled receptor 143 (GPR143) gene were determined by DNA sequencing assays and polymerase chain reaction assays for deletions; all exon coding sequences, exons at the 5'­ and 3'­ends, and non­coding region sequences of intron splicing were assessed. Within the family, nine male patients exhibited disease occurrence at the age of 0­6 months. All patients presented with different degrees of iris depigmentation, horizontal jerk nystagmus, foveal hypoplasia and reduced visual acuity. The fundus of only one patient exhibited choroid coloboma; in the remaining patients, their fundi exhibited different degrees of irregular retinal depigmentation. The mutation c.360+5G>T in the GPR143 gene was identified in this family. In conclusion, the present study identified the splicing mutation c.360+5G>T in the GPR143 gene in a Chinese family with OA1 and successfully identified the site. To the best of our knowledge, there have been no previous reports regarding this mutation in any major genome databases; therefore, this outcome may enrich the mutation spectrum of the GPR143 gene.


Asunto(s)
Albinismo Ocular/genética , Pueblo Asiatico , Proteínas del Ojo/genética , Familia , Glicoproteínas de Membrana/genética , Mutación , Adolescente , Adulto , Anciano , Albinismo Ocular/metabolismo , Albinismo Ocular/patología , China , Proteínas del Ojo/metabolismo , Femenino , Humanos , Masculino , Glicoproteínas de Membrana/metabolismo , Persona de Mediana Edad
9.
Mol Med Rep ; 20(4): 3519-3526, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31485614

RESUMEN

Cisplatin has been widely used as a conventional treatment for patients with non­small cell lung cancer (NSCLC). However, primary and acquired cisplatin resistances are frequently developed during the treatment of patients with NSCLC, leading to an increased mortality rate. Accumulating evidence demonstrated that aberrantly expressed microRNAs (miRs) are involved in the development of chemoresistance. In the present study, sensitivity of NSCLC cells to cisplatin was identified to increase following overexpression of miR­608. Conversely, sensitivity to cisplatin was reduced following miR­608 knockdown. Reverse transcription­quantitative PCR and western blotting analyses identified that TEA domain transcription factor 2 (TEAD2), a key regulator of cell stemness, was negatively regulated by miR­608 in NSCLC cells. By repressing TEAD2, miR­608 decreased the expression level of several target genes of the Hippo­yes­associated protein signaling pathway. Furthermore, TEAD2 mRNA was confirmed to be targeted by miR­608 in NSCLC cells via a dual­luciferase reporter assay. Importantly, the increased cisplatin sensitivity induced by miR­608 overexpression was reversed by transfection of TEAD2 in NSCLC cells. The present data suggested that miR­608 may represent a novel candidate biomarker for the evaluation of cisplatin sensitivity in patients with NSCLC.


Asunto(s)
Biomarcadores de Tumor , Carcinoma de Pulmón de Células no Pequeñas , Cisplatino/farmacología , Proteínas de Unión al ADN , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Pulmonares , MicroARNs , Proteínas de Neoplasias , ARN Neoplásico , Factores de Transcripción , Células A549 , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , MicroARNs/biosíntesis , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Factores de Transcripción de Dominio TEA , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
10.
Oncol Lett ; 10(5): 2802-2806, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26722245

RESUMEN

The present study aimed to investigate the anti-tumor mechanisms of gambogic acid (GA) on NCI-H1993 xenografts in vivo. Non-small cell lung carcinoma NCI-H1993 cells, which harbor a MET gene amplification, were subcutaneously injected into athymic nude mice. The mice were randomly assigned to treatment with 10, 20 or 30 mg/kg GA for 3 weeks. At the end of the efficacy study, all the mice were sacrificed and the tumor tissues were subjected to western blot analysis and immunohistochemical (IHC) staining. GA inhibited NCI-H1993 xenograft tumor growth in a dose-dependent manner. Western blot analysis demonstrated that expression of phosphorylated (p)-MET and its downstream signaling molecules p-AKT and p-ERK1/2 were significantly inhibited by GA. IHC analysis of Ki-67 expression demonstrated that GA treatment resulted in dose-dependent inhibition of tumor cell proliferation. GA exerted antitumor effects on NCI-H1993 xenografts in vivo by direct regulation of the MET signaling pathway. Theses antitumor effects were primarily a result of its anti-proliferation function.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...