Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Front Microbiol ; 12: 667782, 2021.
Article in English | MEDLINE | ID: mdl-34122381

ABSTRACT

Escherichia coli is one major cause of bacterial infections and can horizontally acquire antimicrobial resistance and virulence genes through conjugation. Because conjugative plasmids can rapidly spread among bacteria of different species, the plasmids carrying both antimicrobial resistance and virulence genes may pose a significant threat to public health. Therefore, the identification and characterization of these plasmids may facilitate a better understanding of E. coli pathogenesis and the development of new strategies against E. coli infections. Because iron uptake ability is a potential virulence trait of bacteria, we screened for E. coli conjugative plasmids able to confer both iron uptake ability and ampicillin resistance. The plasmid pEC41, which was derived from the bacteremia clinical isolate EC41, was identified. EC41, which carried the fimH27 allele, belonged to sequence type (ST) 405 and phylogroup D. According to the sequencing analyses, pEC41 was 86 kb in size, and its backbone structure was almost identical to that of another highly conjugative plasmid, pCTX-M3, in which the extended-spectrum ß-lactamase gene bla CTX-M-3 was originally identified. pEC41 carried bla CTX-M-3 and bla TEM-1. The ferric citrate uptake (fec) system was identified in pEC41 and was responsible for conferring iron uptake ability. The fec system contributes to the pathogenesis of EC41 in systemic infections but not in urinary tract infections (UTIs). However, this system promoted competitive fitness of a cystitis-associated clinical isolate to colonize urinary tracts. Additionally, the distribution of the fec system was related to E. coli isolates associated with human bacteremia and UTIs. In summary, the present study identified a novel conjugative plasmid, pEC41, which conferred both antimicrobial resistance and an extra iron uptake ability to E. coli. The iron uptake ability was encoded in the fec system and contributed to E. coli pathogenesis. This study is the first to show that the fec system is a virulence factor in E. coli.

2.
J Exp Clin Cancer Res ; 40(1): 95, 2021 Mar 13.
Article in English | MEDLINE | ID: mdl-33712045

ABSTRACT

BACKGROUND: Ectopic insulin-like growth factor binding protein 3 (IGFBP3) expression has been shown to enhance cell migration and lymph node metastasis of oral squamous cell carcinoma (OSCC) cells. However, OSCC patients with high IGFBP3 expression had improved survival compared with those with low expression. Therefore, we speculated that IGFBP3 expression may play a role in response to conventional OSCC therapies, such as radiotherapy. METHODS: We used in vitro and in vivo analyses to explore IGFBP3-mediated radiosensitivity. Reactive oxygen species (ROS) detection by flow cytometry was used to confirm IGFBP3-mediated ionizing radiation (IR)-induced apoptosis. Geneset enrichment analysis (GSEA) and ingenuity pathway analysis (IPA) were used to analyze the relationship between IGFBP3 and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling. Assays involving an NF-κB inhibitor, ROS scavenger or interleukin 6 (IL-6) were used to evaluate the NF-κB/IL-6/ROS signaling in IGFBP3-mediated radiosensitivity. RESULTS: Ectopic IGFBP3 expression enhanced IR-induced cell-killing in vitro. In vivo, IGFBP3 reduced tumor growth and increased apoptotic signals of tumor tissues in immunocompromised mice treated with IR. Combined with IR, ectopic IGFBP3 expression induced mitochondria-dependent apoptosis, which was apparent through mitochondrial destruction and increased ROS production. Ectopic IGFBP3 expression enhanced NK-κB activation and downstream cytokine expression. After IR exposure, IGFBP3-induced NF-κB activation was inhibited by the ROS scavenger N-acetyl-L-cysteine (NAC). IGFBP3-mediated ROS production was reduced by the NF-κB inhibitor BMS-345541, while exogenous IL-6 rescued the NF-κB-inhibited, IGFBP3-mediated ROS production. CONCLUSIONS: Our data demonstrate that IGFBP3, a potential biomarker for radiosensitivity, promotes IR-mediated OSCC cell death by increasing ROS production through NF-κB activation and cytokine production.


Subject(s)
Carcinoma, Squamous Cell/genetics , Insulin-Like Growth Factor Binding Protein 3/metabolism , Mouth Neoplasms/genetics , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Animals , Apoptosis , Carcinoma, Squamous Cell/pathology , Humans , Male , Mice , Mice, Nude , Mouth Neoplasms/pathology , Signal Transduction
3.
Sci Rep ; 11(1): 644, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436772

ABSTRACT

Ephrin type-A receptor 10 (EPHA10) has been implicated as a potential target for breast and prostate cancer therapy. However, its involvement in oral squamous cell carcinoma (OSCC) remains unclear. We demonstrated that EPHA10 supports in vivo tumor growth and lymphatic metastasis of OSCC cells. OSCC cell migration, epithelial mesenchymal transition (EMT), and sphere formation were found to be regulated by EPHA10, and EPHA10 was found to drive expression of some EMT- and stemness-associated transcription factors. Among EPHA10 ligands, exogenous ephrin A4 (EFNA4) induced the most OSCC cell migration and sphere formation, as well as up-regulation of SNAIL, NANOG, and OCT4. These effects were abolished by extracellular signal-regulated kinase (ERK) inhibition and NANOG knockdown. Also, EPHA10 was required for EFNA4-induced cell migration, sphere formation, and expression of NANOG and OCT4 mRNA. Our microarray dataset revealed that EFNA4 mRNA expression was associated with expression of NANOG and OCT4 mRNA, and OSCC patients showing high co-expression of EFNA4 with NANOG or OCT4 mRNA demonstrated poor recurrence-free survival rates. Targeting forward signaling of the EFNA4-EPHA10 axis may be a promising therapeutic approach for oral malignancies, and the combination of EFNA4 mRNA and downstream gene expression may be a useful prognostic biomarker for OSCC.


Subject(s)
Cell Movement , Ephrin-A4/metabolism , Gene Expression Regulation, Neoplastic , Mouth Neoplasms/pathology , Nanog Homeobox Protein/metabolism , Receptors, Eph Family/metabolism , Spheroids, Cellular/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Ephrin-A4/genetics , Epithelial-Mesenchymal Transition , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Nanog Homeobox Protein/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prognosis , Receptors, Eph Family/genetics , Spheroids, Cellular/metabolism , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Cancers (Basel) ; 12(1)2019 Dec 24.
Article in English | MEDLINE | ID: mdl-31878324

ABSTRACT

We established the NHRI-HN1 cell line from a mouse tongue tumor induced by 4-nitroquinoline 1-oxide (4-NQO)/arecoline, with further selection for cell stemness via in vitro sphere culture, to evaluate potential immunotherapies for oral squamous cell carcinoma (OSCC) in East and Southeast Asia. In vivo and in vitro phenotypic characterization, including tumor growth, immune modulator administration, gene expression, morphology, migration, invasion, and sphere formation assays, were conducted. NHRI-HN1 cells are capable of generating orthotopic tumors in syngeneic mice. Interestingly, immune stimulation via CpG oligodeoxynucleotide (CpG-ODN) dramatically reduced the tumor growth in NHRI-HN1 cell-injected syngeneic mice. The pathways enriched in genes that were differentially expressed in NHRI-HN1 cells when compared to non-tumorigenic cells were similar to those that were identified when comparing human OSCC and non-tumorous tissues. NHRI-HN1 cells have characteristics of epithelial-mesenchymal transition (EMT), including enhanced migration and invasion. NHRI-HN1 cells showed aggressive cell growth and sphere formation. The blockage of extracellular signal-regulated kinase (ERK) activation suppressed cell migration and reduced stemness characteristics in NHRI-HN1 cells, similar to human OSCC cell lines. Our data suggest that NHRI-HN1 cells, showing tumorigenic characteristics of EMT, cancer stemness, and ERK activation, are sufficient in modeling human OSCC and also competent for use in investigating oral cancer immunotherapies.

5.
J Vis Exp ; (151)2019 09 27.
Article in English | MEDLINE | ID: mdl-31609349

ABSTRACT

Cell co-culture assays have been widely used for studying cell-cell interactions between different cell types to better understand the biology of diseases including cancer. However, it is challenging to clarify the complex mechanism of intercellular interactions in highly heterogeneous cell populations using conventional co-culture systems because the heterogeneity of the cell subpopulation is obscured by the average values; the conventional co-culture systems can only be used to describe the population signal, but are incapable of tracking individual cells behavior. Furthermore, conventional single-cell experimental methods have low efficiency in cell manipulation because of the Poisson distribution. Microfabricated devices are an emerging technology for single-cell studies because they can accurately manipulate single cells at high-throughput and can reduce sample and reagent consumption. Here, we describe the concept and application of a microfluidic chip for multiple single-cell co-cultures. The chip can efficiently capture multiple types of single cells in a culture chamber (~46%) and has a sufficient culture space useful to study the cells' behavior (e.g., migration, proliferation, etc.) under cell-cell interaction at the single-cell level. Lymphatic endothelial cells and oral squamous cell carcinoma were used to perform a single-cell co-culture experiment on the microfluidic platform for live multiple single-cell interaction studies.


Subject(s)
Cell Communication , Coculture Techniques/instrumentation , Coculture Techniques/methods , Endothelial Cells/cytology , Microfluidic Analytical Techniques/instrumentation , Mouth Neoplasms/pathology , Single-Cell Analysis/methods , Carcinoma, Squamous Cell/pathology , Cell Movement , Cell Proliferation , Cells, Cultured , Humans , Lab-On-A-Chip Devices
6.
Lab Chip ; 19(8): 1370-1377, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30888367

ABSTRACT

Studies on cellular heterogeneity have emerged as a powerful approach for developing new strategies to treat diseases including cancer. However, it is difficult to set up an in vitro co-culture experiment to study the interaction of individual live cells. In this paper, we report a hydrodynamic shuttling chip (HSC) which can deterministically capture single cells into microfluidic chambers to set up multiple single-cell co-culture experiments in which individual live cells can be microscopically observed. Using this chip device, we demonstrated a triple single-cell culture of oral squamous cell carcinoma and lymphatic endothelial cells to observe the effect of cell-cell interaction on the cell motility. Triple, single-cell pairing efficiency by our HSC device was eightfold higher than that of the probabilistic method. Using this HSC device, we were able to perform triple-culture experiments to show the cell type-dependent motility of oral squamous cell carcinoma and lymphatic endothelial cells, which was not observed in co-culture experiments.


Subject(s)
Cell Separation/instrumentation , Hydrodynamics , Lab-On-A-Chip Devices , Single-Cell Analysis/instrumentation , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Endothelial Cells/cytology , Equipment Design , Humans , Mouth Neoplasms/pathology
7.
Oncogene ; 38(23): 4480-4495, 2019 06.
Article in English | MEDLINE | ID: mdl-30765861

ABSTRACT

In an effort to understand the underlying mechanisms of lymph node metastasis in oral squamous cell carcinoma (OSCC), through in vivo selection, LN1-1 cells were previously established from OEC-M1 cells and showed enhanced lymphangiogenesis and lymphatic metastasis capabilities. In the current study, we use a stable isotope labeling with amino acids in cell culture (SILAC) and liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based proteomic platform to compare LN1-1 to OEC-M1 cells. Interferon-stimulated gene 15 (ISG15) was found highly expressed in LN1-1 cells. Immunohistochemical analysis and meta-analysis of publicly available microarray datasets revealed that the ISG15 level was increased in human OSCC tissues and associated with poor disease outcome. Knockdown of ISG15 had minimal effects on tumor growth but did decrease tumor lymphangiogenesis and lymphatic metastasis of LN1-1 cells. Consistent with the in vivo assay, ISG15 knockdown did not impair cell growth but diminished cell migration, invasion, and transendothelial migration in vitro. ISG15-induced cell migration was independent of ISGylation and associated with membrane protrusion. Ectopic expression of ISG15 increased Rac1 activity and knockdown of Rac1 impaired ISG15-enhanced migration. Furthermore, Rac1 colocalized with ISG15 to a region of membrane protrusion and ISG15 coimmunoprecipitated with Rac1, especially with the Rac1-GDP form. Importantly, as shown by proximity ligation assays, ISG15 and Rac1 physically interacted with each other. Our results indicated that ISG15 affects cell migration by interacting with Rac1 and regulating Rac1 activity and contributes to lymphatic metastasis in OSCC.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cytokines/metabolism , Lymphatic Metastasis , Mouth Neoplasms/pathology , Ubiquitins/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Keratinocytes/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms/metabolism , Neoplasm Invasiveness , Prognosis , Proteomics
8.
Int J Cancer ; 144(11): 2795-2810, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30485433

ABSTRACT

Oral squamous cell carcinoma (OSCC) LN1-1 cells previously showed greater capacities for lymphangiogenesis and lymph node metastasis compared to their parental OEC-M1 cells, in addition to an ability to enhance the migration and tube formation of lymphatic endothelial cells (LECs). Purified by a series of differential centrifugations and characterized using electron microscopy, dynamic light scattering and western blot, LN1-1 cell-derived extracellular vesicles (LN1-1 EVs) were shown to promote LEC migration, tube formation and uptake by LECs more effectively than did OEC-M1 cell-derived EVs (OEC-M1 EVs). Using stable isotope labeling with amino acids in cell culture/liquid chromatography-tandem mass spectrometry-based proteomic platform, the laminin-332 proteins, including laminin α3, ß3 and γ2, were validated as highly expressed proteins in LN1-1 EVs. Clinically, a higher level of laminin-332 was detected in plasma EVs from OSCC patients with lymph node metastasis than in both healthy controls and OSCC patients without lymphatic metastasis, suggesting EV-borne laminin-332 as a novel and noninvasive biomarker for the detection of lymph node metastasis in OSCC. The knockdown of laminin γ2 and inhibition by anti-laminin-332 neutralizing antibodies impaired LN1-1 EV-mediated LEC migration, tube formation and uptake by LECs. Importantly, laminin γ2-deficient EVs showed a reduced ability to drain into lymph nodes in comparison with the control EVs. In addition, the laminin 332/γ2-mediated EV uptake was dependent on integrin α3 but not ß1, ß4 or α6. Collectively, the uptake of laminin γ2-enriched EVs by LECs enhanced in vitro lymphangiogenesis and EV-borne laminin-332 is thus a viable biomarker for OSCC.


Subject(s)
Integrin alpha3/metabolism , Laminin/metabolism , Lymphangiogenesis , Mouth Neoplasms/pathology , Squamous Cell Carcinoma of Head and Neck/pathology , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Endothelial Cells/pathology , Extracellular Vesicles/pathology , Gene Knockdown Techniques , Humans , Laminin/genetics , Lymph Nodes/pathology , Lymphatic Metastasis/diagnosis , Lymphatic Metastasis/pathology , Lymphatic Vessels/cytology , Male , Mice , Mice, Nude , Xenograft Model Antitumor Assays
10.
Sci Rep ; 6: 37450, 2016 11 24.
Article in English | MEDLINE | ID: mdl-27881867

ABSTRACT

Mobilisation of endothelial progenitor cells (EPCs) from the bone marrow is a crucial step in the formation of de novo blood vessels, and levels of peripheral blood EPCs have been shown to be elevated in certain malignant states. Using flow cytometry and a Hill-based colony forming unit (CFU) assay, the present study indicated that higher levels of CD34 and vascular endothelial growth factor receptor 2 (VEGFR2) double-positive EPCs, as well as increased formation of endothelial cell colony-forming units (EC-CFUs) are associated with benign and malignant breast diseases, providing possible indicators for breast disease detection. Gene expression profiles revealed a genetic difference between CD34+ VEGFR2+ EPCs and EC-CFUs. Decreased expression of tumour necrosis factor receptor 2 (TNFR2) signalling-related genes and inhibition of tumour necrosis factor (TNF)-induced signalling were demonstrated in EC-CFUs derived from patients with malignant breast disease in comparison with those from healthy controls. Interestingly, our data provided the first evidence that EC-CFUs derived from patients with malignant breast disease were resistant to TNF-α-induced apoptosis, indicating a plausible target for future therapeutic interventions.


Subject(s)
Breast Neoplasms/diagnosis , Carcinoma, Intraductal, Noninfiltrating/diagnosis , Carcinoma, Lobular/diagnosis , Carcinoma, Papillary/diagnosis , Endothelial Progenitor Cells/metabolism , Gene Expression Regulation, Neoplastic , Hyperplasia/diagnosis , Adult , Antigens, CD34/genetics , Antigens, CD34/metabolism , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/metabolism , Carcinoma, Intraductal, Noninfiltrating/pathology , Carcinoma, Lobular/genetics , Carcinoma, Lobular/metabolism , Carcinoma, Lobular/pathology , Carcinoma, Papillary/genetics , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Case-Control Studies , Colony-Forming Units Assay , Diagnosis, Differential , Disease Resistance/genetics , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/pathology , Female , Gene Expression Profiling , Humans , Hyperplasia/genetics , Hyperplasia/metabolism , Hyperplasia/pathology , Middle Aged , Primary Cell Culture , Receptors, Tumor Necrosis Factor, Type II/genetics , Receptors, Tumor Necrosis Factor, Type II/metabolism , Signal Transduction , Stem Cells/metabolism , Stem Cells/pathology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
12.
Oncotarget ; 6(39): 41837-55, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26540630

ABSTRACT

Frequent metastasis to the cervical lymph nodes leads to poor survival of patients with oral squamous cell carcinoma (OSCC). To understand the underlying mechanisms of lymph node metastasis, two sublines were successfully isolated from cervical lymph nodes of nude mice through in vivo selection, and identified as originating from poorly metastatic parental cells. These two sublines specifically metastasized to cervical lymph nodes in 83% of mice, whereas OEC-M1 cells did not metastasize after injection into the oral cavity. After gene expression analysis, we identified insulin-like growth factor binding protein 3 (IGFBP3) as one of the significantly up-regulated genes in the sublines in comparison with their parental cells. Consistently, meta-analysis of the public microarray datasets and IGFBP3 immunohistochemical analysis revealed increased both levels of IGFBP3 mRNA and protein in human OSCC tissues when compared to normal oral or adjacent nontumorous tissues. Interestingly, the up-regulated IGFBP3 mRNA expression was significantly associated with OSCC patients with lymph node metastasis. IGFBP3 knockdown in the sublines impaired and ectopic IGFBP3 expression in the parental cells promoted migration, transendothelial migration and lymph node metastasis of orthotopic transplantation. Additionally, ectopic expression of IGFBP3 with an IGF-binding defect sustained the IGFBP3-enhanced biological functions. Results indicated that IGFBP3 regulates metastasis-related functions of OSCC cells through an IGF-independent mechanism. Furthermore, exogenous IGFBP3 was sufficient to induce cell motility and extracellular signal-regulated kinase (ERK) activation. The silencing of integrin ß1 was able to impair exogenous IGFBP3-mediated migration and ERK phosphorylation, suggesting a critical role of integrin ß1 in IGFBP3-enchanced functions.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cell Movement , Head and Neck Neoplasms/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Integrin beta1/metabolism , Mouth Neoplasms/metabolism , Somatomedins/metabolism , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/secondary , Cell Line, Tumor , Databases, Genetic , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Insulin-Like Growth Factor Binding Protein 3/genetics , Integrin beta1/genetics , Lymphatic Metastasis , Male , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Phosphorylation , RNA Interference , Signal Transduction , Squamous Cell Carcinoma of Head and Neck , Time Factors , Transfection
13.
Electrophoresis ; 36(17): 2027-34, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26171676

ABSTRACT

The metastatic status of oral cancer is highly associated with the overall survival rate of patients. Previous studies have revealed that the endogenous tryptophan metabolite 5-methoxytryptophan (5-MTP) can downregulate cyclooxygenase-2 expression; suppress tumor proliferation, migration, and invasion; and reduce the tumor size. To improve the understanding of the molecular mechanisms involved in the regulation of 5-MTP in the tumorigenesis of oral cancer, we conducted a comparative wound healing and transwell invasion assays. Our results revealed that 5-MTP reduce oral cancer cell migration and invasion ability. In addition, the results of an in vivo assay demonstrated that the growth of primary tumors was significantly inhibited by 5-MTP in OC3 oral cancer cells and in invasive OC3-I5 oral cancer cells. Moreover, enlarged spleens were observed in OC3-I5-implanted severe combined immunodeficiency mice although 5-MTP can inhibit spleen enlargement. Through comparative proteomics, we identified 32 differentially regulated protein spots by using 2D-DIGE/MALDI-TOF MS analyses. Some of the differentially regulated proteins such as amadillo-repeat-containing X-linked protein 1, phosphoglycerate kinase 1, tropomyosin alpha-1, and tropomyosin alpha-4 may be associated with the 5-MTP-dependent inhibition of oral cancer growth and metastasis. We conclude that 5-MTP plays a crucial role in inhibiting in vitro and in vivo cancer invasion and metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/pathology , Mouth Neoplasms/pathology , Proteome/drug effects , Tryptophan/analogs & derivatives , Animals , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Female , Humans , Mice , Mice, Nude , Mouth Neoplasms/metabolism , Neoplasm Invasiveness , Proteome/analysis , Proteomics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tryptophan/pharmacology , Two-Dimensional Difference Gel Electrophoresis
14.
Electrophoresis ; 36(6): 902-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25545859

ABSTRACT

Diabetic retinopathy typically causes poor vision and blindness. A previous study revealed that a high blood glucose concentration induces glycoxidation and weakens the retinal capillaries. Nevertheless, the molecular mechanisms underlying the effects of high blood glucose induced diabetic retinopathy remain to be elucidated. In the present study, we cultured the retinal pigmented epithelial cell line ARPE-19 in mannitol-balanced 5.5, 25, and 100 mM glucose media and investigated protein level alterations. Proteomic analysis revealed significant changes in 137 protein features, of which 124 demonstrated changes in a glucose concentration dependent manner. Several proteins functionally associated with redox regulation, protein folding, or the cytoskeleton are affected by increased glucose concentrations. Additional analyses also revealed that cellular oxidative stress, including endoplasmic reticulum stress, was significantly increased after treatment with high glucose concentrations. However, the mitochondrial membrane potential and cell survival remained unchanged during treatment with high glucose concentrations. To summarize, in this study, we used a comprehensive retinal pigmented epithelial cell based proteomic approach for identifying changes in protein expression associated retinal markers induced by high glucose concentrations. Our results revealed that a high glucose condition can induce cellular oxidative stress and modulate the levels of proteins with functions in redox regulation, protein folding, and cytoskeleton regulation; however, cell viability and mitochondrial integrity are not significantly disturbed under these high glucose conditions.


Subject(s)
Eye Proteins/analysis , Glucose/pharmacology , Proteome/analysis , Proteome/drug effects , Retinal Pigment Epithelium , Cell Line , Electrophoresis, Gel, Two-Dimensional , Eye Proteins/chemistry , Eye Proteins/metabolism , Humans , Oxidation-Reduction , Protein Folding , Retinal Pigment Epithelium/chemistry , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
15.
Environ Mol Mutagen ; 54(6): 429-49, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23813585

ABSTRACT

Within ultraviolet radiation, ultraviolet B (UVB) is the most energetic and damaging to humans. At the protein level, UVB irradiation downregulates the expression of antioxidant enzymes leading to the accumulation of reactive oxygen species (ROS). Due to lacking of a global analysis of UVB-modulated corneal proteome, we investigate in vitro the mechanism of UVB-induced corneal damage to determine whether hyaluronic acid (HA) is able to reduce UVB irradiation-induced injury in human corneal epithelial cells. Accordingly, human corneal epithelial cell lines (HCE-2) were irradiated with UVB, followed by incubation with low molecular weight HA (LMW-HA, 100 kDa) or high molecular weight HA (HMW-HA, 1,000 kDa) to investigate the physiologic protection of HMW-HA in UVB-induced corneal injury, and to perform a global proteomic analysis. The data demonstrated that HA treatment protects corneal epithelial cells in the UVB-induced wound model, and that the molecular weight of HA is a crucial factor. Only HMW-HA significantly reduces the UVB-induced cytotoxic effects in corneal cells and increases cell migration and wound-healing ability. In addition, proteomic analysis showed that HMW-HA might modulate cytoskeleton regulation, signal transduction, biosynthesis, redox regulation, and protein folding to stimulate wound healing and to prevent these UVB-damaged cells from cell death. Further studies evidenced membrane-associated progesterone receptor component 1 (mPR) and malate dehydrogenase (MDH2) play essential roles in protecting corneal cells from UVB irradiation. This study reports on UVB-modulated cellular proteins that might play an important role in UVB-induced corneal cell injury and show HMW-HA to be a potential substance for protecting corneal cells from UVB-induced injury.


Subject(s)
Epithelium, Corneal , Hyaluronic Acid/pharmacology , Malate Dehydrogenase/genetics , Receptors, Progesterone/genetics , Ultraviolet Rays/adverse effects , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cornea/drug effects , Cornea/radiation effects , Cytoprotection , Cytoskeleton/drug effects , Cytoskeleton/radiation effects , Epithelial Cells/drug effects , Epithelial Cells/radiation effects , Epithelium, Corneal/cytology , Epithelium, Corneal/drug effects , Epithelium, Corneal/radiation effects , Humans , Malate Dehydrogenase/metabolism , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Proteomics , RNA Interference , RNA, Small Interfering , Reactive Oxygen Species/radiation effects , Receptors, Progesterone/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects , Wound Healing/drug effects , Wound Healing/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL