Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Open Med (Wars) ; 18(1): 20230665, 2023.
Article in English | MEDLINE | ID: mdl-36910848

ABSTRACT

Laryngeal squamous cell carcinoma (LSCC) is an aggressive and lethal malignant neoplasm with extremely poor prognoses. Accumulating evidence has indicated that preferentially expressed antigen in melanoma (PRAME) is correlated with several kinds of cancers. However, there is little direct evidence to substantiate the biological function of PRAME in LSCC. The purpose of the current study is to explore the oncogenic role of PRAME in LSCC. PRAME expression was analyzed in 57 pairs of LSCC tumor tissue samples through quantitative real-time PCR, and the correlation between PRAME and clinicopathological features was analyzed. The result indicated that PRAME was overexpressed in the LSCC patients and correlated with the TNM staging and lymphatic metastasis. The biological functions and molecular mechanism of PRAME in LSCC progression were investigated through in vitro and in vivo assays. Functional studies confirmed that PRAME facilitated the proliferation, invasion, migration, and epithelial-mesenchymal transition of LSCC cells, and PRAME also promoted tumor growth in vivo. HDAC5 was identified as an upstream regulator that can affect the expression of PRAME. Moreover, PRAME played the role at least partially by activating PI3K/AKT/mTOR pathways. The above findings elucidate that PRAME may be a valuable oncogene target, contributing to the diagnosis and therapy of LSCC.

2.
Neoplasma ; 69(4): 841-858, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35532293

ABSTRACT

The present study aimed to investigate LINC00278 expression in laryngeal squamous cell carcinoma (LSCC) and its involvement in the process of proliferation, migration, and invasion, providing a rationale for mining potential diagnostic and therapeutic targets of LSCC. Univariate and multivariate Cox regression analyses were performed to identify optimal prognostic lncRNAs. MTS, colony formation, wound healing, and Transwell invasion assays were used to determine the effects of LINC00278 overexpression on the proliferation, migration, and invasion of cancer cells. The expressions of signaling pathway-related proteins and epithelial-mesenchymal transition (EMT) marker proteins were detected using western blot. The chromatin immunoprecipitation (ChIP) and dual-luciferase reporter assays were performed to demonstrate the binding of ETS proto-oncogene 1, transcription factor (ETS1), and LINC00278 promoter region. The molecular targets of LINC00278 were identified by RNA sequencing analysis and co-expression analysis. Kaplan-Meier analysis and CIBERSORT algorithm were used to analyze survival and immune cell infiltration based on LINC00278, COL4A1, and COL4A2. Multivariate Cox regression was used to establish a six-gene prognostic model. LINC00278 expression was low in LSCC tissues, and it was significantly associated with the TNM (tumors/nodes/metastases) stage (p<0.001), lymphatic metastasis (p<0.01), and pathological differentiation (p<0.01). LINC00278 overexpression significantly reduced LSCC cell proliferation, migration, and invasion in TU686, TU177, and AMC-HN-8 cell lines. E-cadherin protein expression was increased, while N-cadherin, Vimentin, Zeb1, and Snail protein expression was decreased in the LINC00278 group, compared to the pcDNA3.1 group. Additionally, in AMC-HN-8 and FaDu cell lines, the LINC00278-treated group had significantly lower p-AKT and p-mTOR protein levels than the control group. ETS1 is a direct transcriptional regulator of the LINC00278 gene based on luciferase reporter assays and ChIP experiments. Western blot analysis demonstrated that high LINC00278 expression inhibited both ETS1 expression and phosphorylation. COL4A1/COL4A2 were identified as potential downstream targets of LINC00278. Meanwhile, the LINC00278/COL4A1/COL4A2-dominated low-risk group showed higher antigen-presenting activity and a higher immune score than the high-risk group. The findings indicated that ETS1 upregulated LINC00278 expression on the Y chromosome, which in turn inhibited LSCC growth in vivo and in vitro by inhibiting the AKT/mTOR signaling pathway via downregulation of COL4A1/COL4A2.


Subject(s)
Head and Neck Neoplasms , Laryngeal Neoplasms , Cell Line, Tumor , Cell Movement , Cell Proliferation , Collagen Type IV/genetics , Collagen Type IV/metabolism , Epithelial-Mesenchymal Transition , Feedback , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Humans , Laryngeal Neoplasms/genetics , Laryngeal Neoplasms/pathology , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics
3.
Anal Bioanal Chem ; 414(17): 4953-4962, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35567611

ABSTRACT

The improper use of phosgene, either as a chemical warfare agent or a leak during chemical production, causes significant risks to human life and property. Therefore, it is particularly important to develop a rapid and highly selective method for the detection of phosgene. In this article, a highly selective fluorescent sensor ONB with a BODIPY unit as a fluorophore and o-aminophenol as a reactive site was constructed for the selective and rapid detection of phosgene in solution. The ONB-containing nanofibers were sprayed onto a non-woven fabric by electrostatic spinning and cut into test films, which can be used well for the detection of gaseous phosgene. While, there were no reported bio-imaging applications for phosgene detection. In this work, nasal mucosa and lung samples from the mice exposed to gaseous phosgene after dropping the ONB solution through the nasal cavity achieved bio-imaging applications successfully.


Subject(s)
Chemical Warfare Agents , Phosgene , Animals , Boron Compounds , Chemical Warfare Agents/toxicity , Gases/chemistry , Lung , Mice , Nasal Mucosa , Phosgene/chemistry , Phosgene/toxicity
4.
Breast Cancer ; 27(5): 828-836, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32279180

ABSTRACT

PURPOSE: Thyroid autoimmunity might be in relation to other autoimmune endocrine disease or non-endocrine disorders and there are innate and adaptive immune cells in breast cancer. Because autoimmune factors are common characteristics of both thyroid autoimmunity and breast cancer, these two types of diseases may occur concurrently in certain patients. The chief goal of this meta-analysis is to perform a combined analysis of the raw data from all included studies, and thereby obtain a reliable conclusion concerning whether TgAb or TPOAb positivity and breast cancer are indeed correlated. METHODS: To determine whether a correlation exists between TgAb or TPOAb positivity and breast cancer, this study performed a review of the literature that began by searching for articles in Chinese or English from the Medline, Embase, Web of Science core, Wanfang, Weipu and SinoMed databases, published during the time span extending from January 1980 to December 2017. On the basis of these raw data, we calculated odds ratio (OR) values, 95% confidence interval (CI) values, and P values. RESULTS: A total of 11 studies were included in this study. By combining the raw data from the retrieved studies, we were able to perform a meta-analysis. The results of this meta-analysis support the hypothesis that patients with breast cancer have a higher TgAb or TPOAb positive rate than the non-breast disease control group (TgAb: OR = 2.71, 95% CI = 1.81-4.05, P < 0.001; TPOAb: OR = 2.86, 95% CI = 2.17-3.77, P < 0.001, respectively). Testing for publication bias indicated that no significant publication bias was present in this meta-analysis, and sensitivity analysis indicated that the results of analysis were stable and reliable. CONCLUSIONS: The results of this meta-analysis suggest strongly that, the TgAb or TPOAb positive rate among patients with breast cancer should be higher than among the non-breast disease control group.


Subject(s)
Autoantibodies/blood , Autoantigens/immunology , Breast Neoplasms/complications , Iodide Peroxidase/immunology , Iron-Binding Proteins/immunology , Thyroiditis, Autoimmune/epidemiology , Autoantibodies/immunology , Breast Neoplasms/blood , Breast Neoplasms/immunology , Female , Humans , Prevalence , Thyroiditis, Autoimmune/blood , Thyroiditis, Autoimmune/diagnosis , Thyroiditis, Autoimmune/immunology
5.
J Asian Nat Prod Res ; 22(6): 588-593, 2020 Jun.
Article in English | MEDLINE | ID: mdl-30982343

ABSTRACT

A new isoflavone glycoside named as 8-O-methylrelusin-7-O-ß-D-apifuranosyl-(1→2)-ß-D-glucopyranoside (1), together with two known compounds, 8-O-methylrelusin-7-O-ß-D-glucopyranoside (2) and isobiflorin (3), were isolated from Abrus cantoniensis. The structure of the new compound was elucidated on the basis of spectroscopic methods including extensive 1D NMR, 2D NMR, and HRESIMS. This is the first report of isoflavone from Abrus cantoniensis. Moreover, all isolated compounds were evaluated for their cytotoxicity against SMMC-7721 and MHCC97-H cell lines.[Formula: see text].


Subject(s)
Abrus , Cardiac Glycosides , Isoflavones , Glycosides , Molecular Structure
6.
Exp Eye Res ; 132: 216-24, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25613074

ABSTRACT

A tissue-engineered human corneal stroma (TE-HCS) has been developed as a promising equivalent to the native corneal stroma for replacement therapy. However, there is still a crucial need to improve the current approaches to render the TE-HCS equivalent more favorable for clinical applications. At the present study, we constructed a TE-HCS by incubating non-transfected human corneal stromal (HCS) cells in an acellular porcine corneal stromata (aPCS) scaffold in 20% fetal bovine serum supplemented DMEM/F12 (1:1) medium at 37 °C with 5% CO2in vitro. After 3 days of incubation, the constructed TE-HCS had a suitable tensile strength for transplantation, and a transparency that is comparable to native cornea. The TE-HCS had a normal histological structure which contained regularly aligned collagen fibers and differentiated HCS cells with positive expression of marker and functional proteins, mimicking a native HCS. After transplantation into rabbit models, the TE-HCS reconstructed normal corneal stroma in vivo and function well in maintaining corneal clarity and thickness, indicating that the completely biological TE-HCS could be used as a HCS equivalent. The constructed TE-HCS has promising potentials in regenerative medicine and treatment of diseases caused by corneal stromal disorders.


Subject(s)
Corneal Diseases/surgery , Corneal Stroma/cytology , Corneal Transplantation/methods , Tissue Engineering/methods , Analysis of Variance , Animals , Cells, Cultured , Corneal Edema/pathology , Disease Models, Animal , Humans , Rabbits , Swine , Tissue Scaffolds
7.
Drug Chem Toxicol ; 38(1): 16-21, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24641202

ABSTRACT

Pranoprofen (PPF), a non-steroidal anti-inflammatory drugs (NSAIDs), is often used in keratitis treatment in clinic. Several studies have assessed in vitro the cytotoxicity of topical NSAIDs to corneal epithelial cells due to its importance for predicting human corneal toxicity. Damage by cytotoxic drugs can result in excessive loss of human corneal endothelial (HCE) cells which lead to decompensation of the endothelium and eventual loss of visual acuity. However, the endothelial cytotoxicity of PPF has not yet been reported using an in vitro model of HCE cells. This study assessed the cytotoxicity of PPF to HCE cells and its underlying mechanism. Cellular viability was determined using inverted phase contrast light microscopy, and plasma membrane permeability, genomic DNA fragmentation, and ultrastructure were detected by acridine orange/ethidium bromide staining, DNA agarose gel electrophoresis, and transmission electron microscopy (TEM), respectively. The results on cellular viability showed that PPF at concentrations ranging from 0.0625 to 1.0 g/l had poignant cytotoxicity to HCE cells, and the extent of its cytotoxicity was dose- and time-dependent. Further characterization indicated that PPF induced plasma membrane permeability elevation, DNA fragmentation, and apoptotic body formation, proving its apoptosis inducing effect on HCE cells. In conclusion, PPF above 0.0625 g/l has poignant cytotoxicity on HCE cells in vitro by inducing cell apoptosis, and should be carefully employed in eye clinic.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Apoptosis/drug effects , Benzopyrans , Endothelial Cells/drug effects , Endothelium, Corneal/drug effects , Propionates , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Benzopyrans/administration & dosage , Benzopyrans/toxicity , Cell Culture Techniques , Cell Line , Cell Membrane Permeability/drug effects , Cell Survival/drug effects , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Endothelial Cells/ultrastructure , Endothelium, Corneal/ultrastructure , Humans , Microscopy, Electron, Transmission , Microscopy, Phase-Contrast , Propionates/administration & dosage , Propionates/toxicity , Time Factors
8.
Int J Ophthalmol ; 7(1): 14-21, 2014.
Article in English | MEDLINE | ID: mdl-24634857

ABSTRACT

AIM: To demonstrate the cytotoxic effect of betaxolol and its underlying mechanism on human corneal endothelial cells (HCE cells) in vitro and cat corneal endothelial cells (CCE cells) in vivo, providing experimental basis for safety anti-glaucoma drug usage in clinic of ophthalmology. METHODS: In vivo and in vitro experiments were conducted to explore whether and how betaxolol participates in corneal endothelial cell injury. The in vitro morphology, growth status, plasma membrane permeability, DNA fragmentation, and ultrastructure of HCE cells treated with 0.021875-0.28g/L betaxolol were examined by light microscope, 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide (MTT) assay, acridine orange (AO)/ethidium bromide (EB) double-fluorescent staining, DNA agarose gel electrophoresis, and transmission electron microscope (TEM). The in vivo density, morphology, and ultrastructure of CCE cells, corneal thickness, and eye pressure of cat eyes treated with 0.28g/L betaxolol were investigated by specular microscopy, applanation tonometer, alizarin red staining, scanning electron microscope (SEM), and TEM. RESULTS: Exposure to betaxolol at doses from 0.0875g/L to 2.8g/L induced morphological and ultrastructural changes of in vitro cultured HCE cells such as cytoplasmic vacuolation, cellular shrinkage, structural disorganization, chromatin condensation, and apoptotic body appearance. Simultaneously, betaxolol elevated plasma membrane permeability and induced DNA fragmentation of these cells in a dose-dependent manner in AO/EB staining. Furthermore, betaxolol at a dose of 2.8g/L also induced decrease of density of CCE cells in vivo, and non-hexagonal and shrunk apoptotic cells were also found in betaxolol-treated cat corneal endothelia. CONCLUSION: Betaxolol has significant cytotoxicity on HCE cells in vitro by inducing apoptosis of these cells, and induced apoptosis of CCE cells in vivo as well. The findings help provide new insight into the apoptosis-inducing effect of anti-glaucoma drugs in eye clinic.

9.
Basic Clin Pharmacol Toxicol ; 114(4): 352-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24373304

ABSTRACT

Lidocaine has been reported to induce apoptosis on rabbit corneal endothelial cells. However, the apoptotic effect and exact mechanism involved in cytotoxicity of lidocaine are not well-established in human corneal endothelial (HCE) cells. In this study, we investigated the apoptosis-inducing effect of lidocaine on HCE cells in vitro. After HCE cells were treated with lidocaine at concentrations of 0.15625-10.0 g/l, the morphology and ultrastructure of the cells were observed by inverted light microscope and transmission electron microscope (TEM). Cell viability was measured by MTT assay, and the apoptotic ratio was evaluated with flow cytometry and fluorescent microscopic counting after FITC-Annexin V/PI and AO/EB staining. DNA fragmentation was detected by electrophoresis, and the activation of caspases was evaluated by ELISA. In addition, changes in mitochondrial membrane potential were determined by JC-1 staining. Results suggest that lidocaine above 1.25 g/l reduced cellular viability and triggered apoptosis in HCE cells in a time- and dose-dependent manner. Diminishment of ΔΨm and the activation of caspases indicate that lidocaine-induced apoptosis was caspase dependent and may be related to mitochondrial pathway.


Subject(s)
Cornea/drug effects , Endothelial Cells/drug effects , Lidocaine/toxicity , Apoptosis/drug effects , Benzimidazoles/metabolism , Carbocyanines/metabolism , Cell Survival/drug effects , Cells, Cultured , Cornea/cytology , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Microscopy, Electron, Transmission , Mitochondria/drug effects
10.
Int J Ophthalmol ; 5(3): 286-92, 2012.
Article in English | MEDLINE | ID: mdl-22773974

ABSTRACT

AIM: To establish an untransfected human corneal stromal (HCS) cell line and characterize its biocompatibility to acellular porcine corneal stroma (aPCS). METHODS: Primary culture was initiated with a pure population of HCS cells in DMEM/F12 media (pH 7.2) containing 20% fetal bovine serum and various necessary growth factors. The established cell line was characterized by growth property, chromosome analysis, tumorigenicity assay, expression of marker proteins and functional proteins. Furthermore, the biocompatibility of HCS cells with aPCS was examined through histological and immunocytochemistry analyses and with light, electron microscopies. RESULTS: HCS cells proliferated to confluence 2 weeks later in primary culture and have been subcultured to passage 140 so far. A continuous untransfected HCS cell line with a population doubling time of 41.44 hours at passage 80 has been determined. Results of chromosome analysis, morphology, combined with the results of expression of marker protein and functional proteins suggested that the cells retained HCS cell properties. Furthermore, HCS cells have no tumorigenicity, and with excellent biocompatibility to aPCS. CONCLUSION: An untransfected and non-tumorigenic HCS cell line has been established, and the cells maintained positive expression of marker proteins and functional proteins. The cell line, with excellent biocompatibility to aPCS, might be used for in vitro reconstruction of tissue-engineered HCS.

SELECTION OF CITATIONS
SEARCH DETAIL
...