Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Publication year range
1.
Discov Oncol ; 14(1): 64, 2023 May 09.
Article in English | MEDLINE | ID: mdl-37160815

ABSTRACT

PURPOSE: Esophageal squamous cell carcinoma (ESCC) remains one of the most common causes of cancer death due to the lack of effective therapeutic options. New targets and the targeted drugs are required to be identified and developed. METHODS: Highly expressed genes in ESCA were identified using the edgeR package from public datasets. Immunostaining assay verified the high expression level of EFNA1 in ESCC. CCK-8, colony formation and wound healing assays were performed to examine the role of EFNA1 and EPHA2 in ESCC progression. Cell cycle was analyzed by flow cytometry and autophagy activation was determined by autophagolysosome formation using transmission electron microscopy. The small molecule targeting to EFNA1 was identified by molecular docking and the anti-tumor effects were verified by in vitro and in vivo models with radiation treatment. RESULTS: EFNA1 was highly expressed in esophageal cancer and significantly associated with poor prognosis. Downregulation of EFNA1 remarkably inhibited cell proliferation and migration. Furthermore, decreased EFNA1 significantly suppressed the expression of cMYC along with its representative downstream genes involved in cell cycle, and activated autophagy. Similar effects on ESCC progression were obtained from knockdown of the corresponding receptor, EPHA2. The potential small molecule targeting to EFNA1, salvianolic acid A (SAA), could significantly suppress ESCC progression and increase the sensitivity to radiotherapy. CONCLUSION: We revealed that EFNA1 facilitated the ESCC progression via the possible mechanism of activating cMYC-modulated cell proliferation and suppressing autophagy, and identified SAA as a potential drug targeting EFNA1, providing new options for the future treatments for ESCC patients.

2.
Oncol Lett ; 22(5): 802, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34630709

ABSTRACT

Laryngeal squamous cell carcinoma (LSCC) is a highly invasive malignant tumor in the head and neck area. As an oncogene, long non-coding RNA (lncRNA) nuclear enriched abundant transcript 1 (NEAT1) promotes cell proliferation, migration and invasion several types of cancer. The present study aimed to reveal the effects of NEAT1 on the progression of LSCC. Reverse transcription-quantitative PCR (RT-qPCR) was used to detect relative mRNA expression levels of NEAT1, microRNA (miR)-204-5p and semaphorin (SEMA) 4B. Kaplan-Meier analysis was used to analyze overall survival times. RNA in-situ hybridization (ISH) exhibited the distribution of NEAT1 and miR-204-5p in tissues. RNA fluorescence ISH was used to analyze the distribution of NEAT1 and miR-204-5p in the cells. Western blot analysis was used to detect the expression level of target proteins. Cell viability was analyzed using a MTT assay, while flow cytometry was used to determine cell apoptosis. Wound healing and Transwell invasion assays were used to value cell migration and invasion, respectively. RNA immunoprecipitation assay, bioinformatics prediction and a dual luciferase reporter assay were used to analyze the target relationship. The RT-qPCR results showed that NEAT1 was highly expressed and miR-204-5p had decreased expression in LSCC tissues and cells compared with that in the normal tissue and the 16HBE-14o cell line, respectively. Knockdown of NEAT1 using small interfering (si) RNA and overexpressed miR-204-5p both effectively inhibited the proliferation, migration and invasion of LSCC cells. Besides, further experiments revealed that miR-204-5p was a target of NEAT1. At the same time, silenced miR-204-5p reversed the anti-tumor effects of si-NEAT1. In addition, SEMA4B was targeted by miR-204-5p in LSCC cells and upregulated SEMA4B weakened the antitumor effects of miR-204-5p in LSCC cells. NEAT1 regulated the expression of SEMA4B by targeting miR-204-5p in LSCC cells. Overall, NEAT1 promoted the proliferation and invasion of LSCC cells by regulating the miR-204-5p/SEMA4B axis.

3.
Mol Biol Rep ; 46(4): 3857-3864, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31066003

ABSTRACT

Although a majority of nasopharyngeal carcinoma (NPC) are undifferentiated and strongly radiosensitive, many NPC patients still have troubles in recurrence. Traditional Chinese medicine (TCM) is considered as potential therapeutic drugs in NPC. However, the effect of Glycyrrhiza glabra on NPC is limited. The present study shows the decreased proliferation and high apoptosis in G. glabra root extract-treated C666-1 cells, indicating the anti-cancerous function of G. glabra in NPC. Then GC/MS-based metabolomics is employed to characterize variation of metabolomes in response to G. glabra root extract treatment. Metabolic category elaborates the higher percentage of down-regulated amino acids and lipids after G. glabra treatment. Moreover, ICA and pathway enrichment analysis further observe that glycine, serine and threonine metabolism, fatty acid biosynthesis, alanine, aspartate and glutamate metabolism, and cysteine and methionine metabolism are four important amino acid and lipid metabolisms that likely contribute to the anti-cancer effect of G. glabra in NPC. These pathways point out the seven metabolite biomarkers, glutathione, glutamine, L-alanine, glycine, L-serine, tetradecanoic acid and stearic acid. Taken together, these findings provide potential clues that anti-cancer mechanisms of G. glabra root extract are linked to the metabolic strategies and emphasize the significance of metabolic strategies against NPC.


Subject(s)
Glycyrrhiza/metabolism , Nasopharyngeal Carcinoma/metabolism , Plant Extracts/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , China , Humans , Medicine, Chinese Traditional/methods , Metabolomics/methods , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Neoplasms/pathology , Neoplasm Recurrence, Local/pathology , Plant Roots/metabolism
4.
J Cell Physiol ; 234(2): 1699-1706, 2019 02.
Article in English | MEDLINE | ID: mdl-30070690

ABSTRACT

Circular RNAs (circRNAs) are reported to regulate the development and progression of multiple cancers. However, the functions of circRNAs in nasopharyngeal carcinoma (NPC) are unclear. In this study, we identified that circular homeodomain interacting protein kinase 3 (circHIPK3) was highly expressed in NPC tissues and cell lines. Moreover, we found that circHIPK3 expression levels could act as a prognostic marker in NPC patients. We showed that circHIPK3 silence repressed NPC cell proliferation, migration, and invasion in vitro. In addition, circHIPK3 depletion dramatically repressed tumor growth and metastasis in vivo. Mechanistically, we revealed circHIPK3 as a competing endogenous RNA of microRNA (miR)-4288 that targets E74-like ETS transcription factor 3 (ELF3) in NPC cells. We found that miR-4288 inhibition reversed the effects of circHIPK3 silence on NPC cells. Furthermore, rescue assays also indicated that circHIPK3 promoted the malignant behaviors of NPC cells via enhancing ELF3 expression by suppressing the miR-4288 levels. In conclusion, our findings demonstrated that circHIPK3 facilitated NPC progression through protecting ELF3 from miR-4288-mediated silencing, which suggested that the circHIPK3-miR-4288-ELF3 regulatory loop might be a potential target for NPC prevention.


Subject(s)
Cell Movement , Cell Proliferation , MicroRNAs/metabolism , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Neoplasms/metabolism , RNA, Circular/metabolism , Adult , Animals , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Male , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Middle Aged , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/secondary , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Neoplasm Invasiveness , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , RNA, Circular/genetics , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Cutan Ocul Toxicol ; 37(3): 245-251, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29480079

ABSTRACT

As a severe photoreceptor-degenerative disease, retinitis pigmentosa (RP) is currently incurable and eventually leads to partial or complete blindness. (3R)-5,6,7-trihydroxy-3-isopropyl-3-methylisochroman-1-one (TIM) is a novel antioxidant isolated from the plant of Alpinia katsumadai Hayata, with protective effects on photoreceptor cells against lipoteichoic acid-induced damage through inhibiting oxidative stress. The present study was to further demonstrate whether TIM could ameliorate retinal degeneration of Pde6brd10 (rd10) mice, a mouse model of RP. rd10 mice were treated with TIM by intraperitoneal injection daily from postnatal Day 10 (P10) to P26. Retinal function was tested by electroretinography. Histology was evaluated by toluidine blue staining and TUNEL assay. Oxidative stress markers were measured by ELISA. Immunohistochemistry, real-time PCR, and western blotting were applied to explore the protective mechanism. Results showed TIM significantly improved the retinal function and decreased photoreceptor cell apoptosis in rd10 mice through reducing oxidative stress. For the first time, this study demonstrated the protective effects of TIM against retinal degeneration in rd10 mice, providing scientific rationale to use TIM treating the RP.


Subject(s)
Alpinia/chemistry , Antioxidants/pharmacology , Chromans/pharmacology , Photoreceptor Cells, Vertebrate/drug effects , Plant Extracts/pharmacology , Retinitis Pigmentosa/drug therapy , Animals , Antioxidants/chemistry , Antioxidants/therapeutic use , Apoptosis/drug effects , Cell Survival , Chromans/chemistry , Chromans/therapeutic use , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Disease Models, Animal , Electroretinography , Humans , Injections, Intraperitoneal , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidative Stress/drug effects , Photoreceptor Cells, Vertebrate/pathology , Plant Extracts/therapeutic use , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology
6.
Mol Med Rep ; 12(2): 2367-73, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25891118

ABSTRACT

MicroRNA (miR)-9 has been demonstrated to regulate the radiosensitivity of tumor cells. In the present study, the mechanism by which miR-9 modulates the sensitivity of nasopharyngeal carcinoma (NPC) cells to ultraviolet (UV) radiation was investigated. The results demonstrated that exposure of NPC cells to UV light resulted in a significant increase in the expression of miR-9, and that CNE2 cells overexpressing miR-9 exhibited reduced levels of DNA damage and increased levels of total glutathione upon UV exposure. Accordingly, the inhibition of the expression of miR-9 promoted UV-induced DNA damage and apoptosis. Although miR-9 inhibited the expression of E-cadherin in the CNE2 cells and increased their resistance to UV radiation, the use of small interfering RNA to inhibit the expression of E-cadherin was not sufficient to decrease the radiosensitivity of the NPC cells. These data demonstrated that miR-9 did not modulate the sensitivity of the CNE2 cells to UV radiation through E-cadherin, but suggested that miR-9 regulated radiosensitivity through its effects on glutathione. These findings suggest that miR-9 may be a potential target for modulating the radiosensitivity of NPC cells.


Subject(s)
Epithelial Cells/radiation effects , Gene Expression Regulation, Neoplastic , Glutathione/metabolism , MicroRNAs/genetics , Radiation Tolerance/genetics , Apoptosis , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Proliferation/radiation effects , DNA Damage , Epithelial Cells/metabolism , Epithelial Cells/pathology , Glutathione/agonists , Glutathione/antagonists & inhibitors , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Nasopharynx/metabolism , Nasopharynx/pathology , Nasopharynx/radiation effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ultraviolet Rays
7.
Am J Rhinol Allergy ; 28(2): 110-6, 2014.
Article in English | MEDLINE | ID: mdl-24717947

ABSTRACT

BACKGROUND: According to the hygiene hypothesis, bacterial infections during early life contribute to a reduced incidence of asthma in animals. However, the effects of microbial products at a safe dose and within a rational time course on the prevention of allergic rhinitis (AR) have been inconclusive. This study investigated the immunomodulatory effects of oral administration of a bacterial extract, OM-85 Broncho-Vaxom (BV), with a low dose and general time course, which is currently used for respiratory infections in humans, on AR inflammation in mice. METHODS: We developed a mouse model of ovalbumin (OVA)-induced AR allergic inflammation in the nose mucosa of mice. Low doses of OM-85 BV were orally administered for 3 months (long term) before sensitization. We evaluated nasal symptoms, pathology in the nose, inflammatory cells, and the levels of T helper 1 (Th1)/Th2 cytokines in the nasal lavage fluids, and the serum levels of specific IgE and IgG1. We also observed enhanced effects of OM-85 BV with 1 month (short term) of treatment. RESULTS: We found that long-term pretreatment with OM-85 BV protected the mice from the majority of allergy-specific symptoms; specifically, OM-85 BV suppressed nasal symptoms, inhibited eosinophil infiltration in the nose, inhibited inflammatory infiltrates and the Th2 response by reducing cytokines (IL-4, IL-5, or IL-13) in the nasal lavage fluids, and reduced IgE and IgG1 levels. Furthermore, short-term treatment with OM-85 BV decreased the levels of Th2 cytokines and IgE. CONCLUSION: Taken together, our data suggested that OM-85 BV is a low-cost alternative candidate to prevent AR with simple oral administration.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens, Bacterial/administration & dosage , Cell Extracts/administration & dosage , Eosinophils/drug effects , Nasal Mucosa/drug effects , Rhinitis, Allergic/prevention & control , Th2 Cells/drug effects , Administration, Oral , Allergens/immunology , Animals , Antibody Formation/drug effects , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Eosinophils/immunology , Female , Humans , Immunoglobulin E/blood , Immunoglobulin G/blood , Immunomodulation , Mice , Mice, Inbred BALB C , Nasal Mucosa/immunology , Ovalbumin/immunology , Rhinitis, Allergic/microbiology , Th1 Cells/drug effects , Th1 Cells/immunology , Th2 Cells/immunology , Time Factors
8.
Article in Chinese | MEDLINE | ID: mdl-24195825

ABSTRACT

OBJECTIVE: To investigate the effects of down-regulated miR-9 expression on ultraviolet rays (UV)-induced reactive oxygen species (ROS) damage in nasopharyngeal carcinoma (NPC) cells. METHODS: The NPC cells were transfected with inhibitors of miR-9 by lipofectamine to decrease the expression of miR-9, and the cells transfected with inhibitor control as the control. ROS levels following UV exposure were examined with DCF-DA method and the concentration of glutathione was analyzed via the benzoic acid method; DNA damage and apoptosis also were evaluated. RESULTS: There was significant difference in ROS levels between miR-9 expression-inhibited cells and control cells (26 895 ± 218 vs 15 765 ± 927, t = 39.754, P < 0.001), and also there were significant differences in DNA damage rates (28.0% ± 10.0% vs 23.6% ± 9.2%) and in apoptosis rates (8.0% ± 0.9% vs 4.5% ± 0.8%) following UV exposure between two groups of cells. The miR-9 expression-inhibited cells showed lower level (1.87 ± 0.15) µmol/L of glutathione compared with the control cells (9.85 ± 0.15) µmol/L (t = -48.832, P < 0.001). CONCLUSION: Inhibition of miR-9 expression promoted UV-induced ROS damage in nasopharyngeal carcinoma cells.


Subject(s)
MicroRNAs/metabolism , Nasopharyngeal Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Apoptosis , Carcinoma , Cell Line, Tumor , DNA Damage , Humans , Nasopharyngeal Carcinoma , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL