Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
J Toxicol Pathol ; 37(1): 39-43, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38283374

ABSTRACT

Ameloblastoma is a locally aggressive tumor derived from the odontogenic epithelium of the developing tooth germ. It is rarely reported in experimental Sprague-Dawley (SD) rats. In this 90-day percutaneous repeated-dose toxicity study, mandibular nodules were observed from day 56 to 90. Upon necropsy, a well-demarcated nodule, approximately 1.2×1.0×1.0 cm, was found attached to the mandibular bone, alongside the unerupted left incisor. Histopathologically, the epithelial cells formed islands, nests, or anastomosing strands. The epithelial islands were surrounded by a peripheral layer of tall columnar cells with antibasilar nuclei arranged in a palisading pattern. Several mitotic cells were observed. Some epithelial islands displayed cystic degenerative changes and squamous metaplasia. Necrotic tissue with inflammatory cell infiltration was observed at the tumor margins. Immunohistochemically, the neoplastic epithelial islands and mesenchymal components exhibited positivity for pan-cytokeratin and vimentin, respectively. Based on these features, the case was diagnosed as follicular ameloblastoma in an SD rat.

2.
Free Radic Biol Med ; 205: 262-274, 2023 08 20.
Article in English | MEDLINE | ID: mdl-37330147

ABSTRACT

Pro-inflammatory cytokines upregulate the expression of the H2O2-producing NADPH oxidase dual oxidase 2 (DUOX2)2 which, when elevated, adversely affects survival from pancreatic ductal adenocarcinoma (PDAC). Because the cGAS-STING pathway is known to initiate pro-inflammatory cytokine expression following uptake of exogenous DNA, we examined whether activation of cGAS-STING could play a role in the generation of reactive oxygen species by PDAC cells. Here, we found that a variety of exogenous DNA species markedly increased the production of cGAMP, the phosphorylation of TBK1 and IRF3, and the translocation of phosphorylated IRF3 into the nucleus, leading to a significant, IRF3-dependent enhancement of DUOX2 expression, and a significant flux of H2O2 in PDAC cells. However, unlike the canonical cGAS-STING pathway, DNA-related DUOX2 upregulation was not mediated by NF-κB. Although exogenous IFN-ß significantly increased Stat1/2-associated DUOX2 expression, intracellular IFN-ß signaling that followed cGAMP or DNA exposure did not itself increase DUOX2 levels. Finally, DUOX2 upregulation subsequent to cGAS-STING activation was accompanied by the enhanced, normoxic expression of HIF-1α and VEGF-A as well as DNA double strand cleavage, suggesting that cGAS-STING signaling may support the development of an oxidative, pro-angiogenic microenvironment that could contribute to the inflammation-related genetic instability of pancreatic cancer.


Subject(s)
Hydrogen Peroxide , Pancreatic Neoplasms , Humans , Dual Oxidases/genetics , Dual Oxidases/metabolism , Hydrogen Peroxide/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Signal Transduction , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , DNA/metabolism , Cytokines , Pancreatic Neoplasms/metabolism , Tumor Microenvironment
3.
Chem Biol Interact ; 380: 110511, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37120125

ABSTRACT

Carteolol is a commonly-used topical medication for primary open-angle glaucoma. However, long-term and frequent ocular application of carteolol entails its residuals at low concentration in the aqueous humor for a long duration and may exert latent toxicity in the human corneal endothelial cells (HCEnCs). Here, we treated the HCEnCs in vitro with 0.0117% carteolol for 10 days. Thereafter, we removed the cartelolol and normally cultured the cells for 25 days to investigate the chronical toxicity of carteolol and the underlying mechanism. The results exhibited that 0.0117% carteolol induces senescent features in the HCEnCs, such as increased senescence-associated ß-galactosidase positive rates, enlarged relative cell area and upregulated p16INK4A and senescence-associated secretory phenotypes, including IL-1α, TGF-ß1, IL-10, TNF-α, CCL-27, IL-6 and IL-8, as well as decreased Lamin B1 expression and cell viability and proliferation. Thereby, further exploration demonstrated that the carteolol activates ß-arrestin-ERK-NOX4 pathway to increase reactive oxygen species (ROS) production that imposes oxidative stress on energetic metabolism causing a vicious cycle between declining ATP and increasing ROS production and downregulation of NAD+ resulting in metabolic disturbance-mediated senescence of the HCEnCs. The excess ROS also impair DNA to activate the DNA damage response (DDR) pathway of ATM-p53-p21WAF1/CIP1 with diminished poly(ADP-Ribose) polymerase (PARP) 1, a NAD+-dependent enzyme for DNA damage repair, resulting in cell cycle arrest and subsequent DDR-mediated senescence. Taken together, carteolol induces excess ROS to trigger HCEnC senescence via metabolic disturbance and DDR pathway.


Subject(s)
Carteolol , Glaucoma, Open-Angle , Humans , Reactive Oxygen Species/metabolism , Cellular Senescence , Signal Transduction/physiology , Endothelial Cells/metabolism , beta-Arrestins/metabolism , NAD/metabolism , NADPH Oxidase 4/metabolism
4.
Nanomaterials (Basel) ; 12(24)2022 Dec 11.
Article in English | MEDLINE | ID: mdl-36558272

ABSTRACT

Ultrathin porous Co3O4 nanosheets are synthesized successfully, the thickness of which is about three unit-cell dimensions. The enhanced oxygen evolution reaction (OER) performance and electronic interaction between Co3O4 and Au is firstly reported in Co3O4 ultrathin porous nanosheets by physically mixing with Au nanoparticles. With the loading of the Au nanoparticles, the current density of ultrathin porous Co3O4 nanosheets is enhanced from 9.97 to 14.76 mA cm-2 at an overpotential of 0.5 V, and the overpotential required for 10 mA cm-2 decreases from 0.51 to 0.46 V, smaller than that of commercial IrO2 (0.54 V). Furthermore, a smaller Tafel slope and excellent durability are also obtained. Raman spectra, XPS measurement, and X-ray absorption near edge structure spectra (XANES) show that the enhanced OER ascribed to a higher Co2+/Co3+ ratio and quicker charge transfer due to the electronic interaction between Au and ultrathin Co3O4 nanosheets with low-coordinated surface, and Co2+ ions are beneficial for the formation of CoOOH active sites.

5.
J Photochem Photobiol B ; 235: 112568, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36137302

ABSTRACT

The human corneal endothelial cells (HCEnCs) play a vital role in the maintenance of corneal transparency and visual acuity. In our daily life, HCEnCs are inevitably exposed to ultraviolet B (UVB) radiation leading to decreases of visual acuity and corneal transparency resulting in visual loss eventually. Therefore, understanding the UVB-induced cytotoxicity in HCEnCs is of importance for making efficient strategies to protect our vision from UVB-damage. However, in-depth knowledge about UVB-induced cytotoxicity in HCEnCs is missing. Herein, we pulse-irradiated the HCEnCs in vitro with 150 mJ/cm2 UVB (the environmental dose) at each subculture for 4 passages to explore the insights into UVB-induced phototoxicity. The results showed that the UVB-treated HCEnCs exhibit typical senescent characteristics, including significantly enlarged relative cell area, increased senescence-associated ß-galactosidase positive staining, and upregulated p16INK4A and senescence associated secretory phenotypes (SASPs) such as CCL-27, IL-1α/6/8/10, TGF-ß1 and TNF-α, as well as decreased cell proliferation and Lamin B1 expression, and translocation of Lamin B1. Furthermore, we explored the causative mechanisms of senescence and found that 150 mJ/cm2 UVB pulse-irradiation impairs DNA to activate DNA damage response (DDR) pathway of ATM-p53-p21WAF1/CIP1 with downregulated DNA repair enzyme PARP1, leading to cell cycle arrest resulting in DDR-mediated senescence. Meanwhile, UVB pulse-irradiation also elicits a consistent increase of ROS production to aggravate DNA damage and impose oxidative stress on energy metabolism leading to metabolic disturbance resulting in metabolic disturbance-mediated senescence. Altogether, the repeated pulse-irradiation of 150 mJ/cm2 UVB induces HCEnC senescence via both DDR pathway and energy metabolism disturbance.


Subject(s)
Cellular Senescence , DNA Damage , Endothelial Cells , Oxidative Stress , Ultraviolet Rays , Cells, Cultured , Cellular Senescence/radiation effects , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Endothelial Cells/cytology , Endothelial Cells/radiation effects , Humans , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays/adverse effects , beta-Galactosidase/metabolism
6.
Life Sci ; 277: 119453, 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-33798551

ABSTRACT

Limbal stem cells (LSCs) are crucial for corneal transparency and vision. Any damages to LSCs might lead to limbal stem cell deficiency resulting in corneal opacification and even blindness. Here, we investigated the cytotoxicity of timolol and its underlying mechanisms in rabbit LSCs (rLSCs) in vitro. High concentrations of 0.5% and 0.25% timolol induced necroptosis in rLSCs to upregulate receptor interacting protein kinase (RIPK)1, RIPK3, mixed lineage kinase domain-like (MLKL) and phosphorylated MLKL along with downregulation of caspase-8 and caspase-2 within 4 h. While, median concentrations of 0.125% to 0.0625% timolol induced apoptosis in the rLSCs within 28 h. The apoptotic mechanism in the median-concentration timolol-treated rLSCs is probably via extrinsic apoptosis pathway by activating caspase-2, caspase-8 and caspase-3 and intrinsic apoptosis pathway triggered by excessive generation of ROS and subsequent DNA damage to upregulate Bax and Bad, downregulate Bcl-2 and Bcl-xL, subsequently disrupt mitochondrial membrane potential, cytosolically translocate cytochrome c and apoptosis-inducing factor, and activate caspase-9. In addition, low concentration of 0.03125% timolol induced senescence in the rLSCs by elevating ROS level and increasing number of senescence associated ß-galactosidase positive cells at 28 h. Our findings reveal that timolol induces necroptosis, apoptosis and senescence concentration-dependently in rLSCs in vitro.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Apoptosis , Cellular Senescence , Limbus Corneae/pathology , Necroptosis , Stem Cells/pathology , Timolol/pharmacology , Animals , In Vitro Techniques , Limbus Corneae/drug effects , Limbus Corneae/metabolism , Male , Phosphorylation , Rabbits , Signal Transduction , Stem Cells/drug effects , Stem Cells/metabolism
7.
PLoS One ; 15(5): e0233208, 2020.
Article in English | MEDLINE | ID: mdl-32428030

ABSTRACT

To facilitate functional investigation of the role of NADPH oxidase 1 (NOX1) and associated reactive oxygen species in cancer cell signaling, we report herein the development and characterization of a novel mouse monoclonal antibody that specifically recognizes the C-terminal region of the NOX1 protein. The antibody was validated in stable NOX1 overexpression and knockout systems, and demonstrates wide applicability for Western blot analysis, confocal microscopy, flow cytometry, and immunohistochemistry. We employed our NOX1 antibody to characterize NOX1 expression in a panel of 30 human colorectal cancer cell lines, and correlated protein expression with NOX1 mRNA expression and superoxide production in a subset of these cells. Although a significant correlation between oncogenic RAS status and NOX1 mRNA levels could not be demonstrated in colon cancer cell lines, RAS mutational status did correlate with NOX1 expression in human colon cancer surgical specimens. Immunohistochemical analysis of a comprehensive set of tissue microarrays comprising over 1,200 formalin-fixed, paraffin-embedded tissue cores from human epithelial tumors and inflammatory disease confirmed that NOX1 is overexpressed in human colon and small intestinal adenocarcinomas, as well as adenomatous polyps, compared to adjacent, uninvolved intestinal mucosae. In contradistinction to prior studies, we did not find evidence of NOX1 overexpression at the protein level in tumors versus histologically normal tissues in prostate, lung, ovarian, or breast carcinomas. This study constitutes the most comprehensive histopathological characterization of NOX1 to date in cellular models of colon cancer and in normal and malignant human tissues using a thoroughly evaluated monoclonal antibody. It also further establishes NOX1 as a clinically relevant therapeutic target in colorectal and small intestinal cancer.


Subject(s)
Adenocarcinoma/enzymology , Colonic Neoplasms/enzymology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Intestine, Small/enzymology , NADPH Oxidase 1/biosynthesis , Neoplasm Proteins/biosynthesis , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Caco-2 Cells , Colonic Neoplasms/genetics , HT29 Cells , Humans , Intestine, Small/pathology , Models, Biological , NADPH Oxidase 1/genetics , Neoplasm Proteins/genetics
8.
Curr Eye Res ; 45(9): 1065-1073, 2020 09.
Article in English | MEDLINE | ID: mdl-32090638

ABSTRACT

PURPOSE: To provide scientific data for clinical practice in making strategies for accelerating corneal endothelial wound healing, we investigated the impact of UVA on the corneal endothelial wound healing process and the underlying mechanism using an in vitro cell model. MATERIALS AND METHODS: An in vitro cell model for corneal endothelial wound healing was established by scratching the in vitro cultured human corneal endothelial cell (HCEnC) confluent layer. Then, we investigated the impacts of UVA irradiation and Ascorbic acid-2-phosphate (Asc-2p) on the wound healing process of the in vitro HCEnC model by examining wound-healing index, F-actin+ rate, Ki-67+ rate, and ROS production. RESULTS: After scratching, the Ki-67+ and F-actin+ HCEnCs occupied the scratching gap. Furthermore, the F-actin+ rates were significantly higher than Ki-67+ rates in the wound closure area. After irradiated with UVA, the wound-healing indexes, Ki-67+ rates and F-actin+ rates of the wound-healing model significantly reduced, whereas the ROS production significantly increased in a dose-dependent manner. Pretreatment with Asc-2p significantly reduced the ROS production as well as increased the wound-healing indexes, Ki-67+rates and F-actin+ rates of the UVA irradiated wound-healing model. CONCLUSION: The migration of HCEnC plays a major role in the wound healing process of the established cell model, which is like the wound healing process in vivo. UVA decreases the wound closure of the in vitro HCEnC model dose-dependently, while antioxidant Asc-2p can attenuate the damage to UVA to HCEnCs probably via reducing ROS to improve their migration.


Subject(s)
Endothelium, Corneal/radiation effects , Ultraviolet Rays , Wound Healing/radiation effects , Actins/metabolism , Ascorbic Acid/analogs & derivatives , Ascorbic Acid/pharmacology , Cell Movement , Cell Proliferation , Cells, Cultured , Endothelium, Corneal/metabolism , Humans , Ki-67 Antigen/metabolism , Models, Biological , Reactive Oxygen Species/metabolism
9.
Naunyn Schmiedebergs Arch Pharmacol ; 393(1): 77-88, 2020 01.
Article in English | MEDLINE | ID: mdl-31420720

ABSTRACT

Norfloxacin, a frequently used ocular antibiotic, might have cytotoxic effect on human corneal endothelial cells (HCECs), subsequently damage the cornea and finally impair human vision. However, the possible mechanisms of cytotoxicity of norfloxacin to HCEC line are unclear. Herein, we investigated the cytotoxicity of norfloxacin and its underlying cellular and molecular mechanisms using in vitro cultured non-transfected HCECs and verified the cytotoxicity with cat corneal endothelium in vivo. In the present study, the cytotoxicity of norfloxacin in the in vitro cultured HCECs was recognized by causing abnormal morphology such as cell shrinkage and detachment from plate bottom, and decline of viability of in vitro cultured HCECs. Then, its cytotoxicity was verified by inducing reduction of cell density and morphological abnormality of in vivo cat corneal endothelial cells. Furthermore, the cytotoxicity of norfloxacin in HCECs was corroborated as apoptosis by elevation of plasma membrane permeability, S phase arrest, phosphatidylserine externalization, DNA fragmentation, and apoptotic body formation in in vitro cultured HCECs and apoptosis-like swollen cells in the in vivo model. Moreover, norfloxacin induced extrinsic death receptor-mediated apoptosis pathway by activating caspase-2/-8/-3 and intrinsic mitochondrion-dependent apoptosis pathway by downregulating anti-apoptotic Bcl-2 and upregulating of pro-apoptotic Bad, which disrupted mitochondrial transmembrane potential, subsequently upregulated cytoplasmic cytochrome c and apoptosis-inducing factor and finally activated caspase-9/-3. Generally, norfloxacin induces HCE cell apoptosis via a death receptor-mediated and mitochondrion-dependent signaling pathway.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cornea/cytology , Endothelial Cells/drug effects , Norfloxacin/pharmacology , Animals , Apoptosis/drug effects , Cats , Cell Cycle/drug effects , Cell Line , Cell Survival/drug effects , DNA Fragmentation , Endothelial Cells/pathology , Endothelial Cells/physiology , Endothelial Cells/ultrastructure , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Microscopy, Electron, Transmission
10.
Toxicology ; 428: 152305, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31605733

ABSTRACT

In the present study, the toxicity of phenylephrine, a selective α1-adrenergic receptor agonist, in corneal epithelial cells and its underlying mechanisms were investigated using an in vitro model of human corneal epithelial cells (HCEPCs) and an in vivo model of New Zealand white rabbit corneas. The HCEPCs treated with phenylephrine at concentrations from 10% to 0.078125% displayed abnormal morphology, decline of cell viability and elevation of plasma membrane permeability time- and dose-dependently. Moreover, 10%-1.25% phenylephrine induce necrosis characteristics of marginalization and uneven distribution of chromatin through up-regulation of RIPK1, RIPK3 and MLKL along with inactivation of caspase-8 and caspase-2, whereas 0.625% phenylephrine induced condensed chromatin, S phase arrest, phosphatidylserine externalization, DNA fragmentation and apoptotic body formation in the HCECs through activation of caspase-2, -8, -9 and -3 as well as down-regulation of Bcl-2, up-regulation of Bad, ΔΨm disruption and release of cytochrome c and AIF into cytosol. At last, 10% phenylephrine induced destruction of the corneal epithelia and apoptosis of corneal epithelial cells in rabbit corneas. In conclusion, 10% to 1.25% phenylephrine cause necroptosis via RIPK1-RIPK3-MLKL axis and 0.625% phenylephrine induce apoptosis via a mitochondrion-dependent and death receptor-mediated signal pathway in HCEPCs.


Subject(s)
Adrenergic alpha-1 Receptor Agonists/toxicity , Apoptosis/drug effects , Epithelial Cells/drug effects , Necroptosis/drug effects , Phenylephrine/toxicity , Animals , Cell Cycle/drug effects , Cell Membrane Permeability/drug effects , Cells, Cultured , Cornea/cytology , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Humans , Male , Rabbits , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Time Factors
11.
J Immunol ; 203(9): 2532-2544, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31548328

ABSTRACT

Dual oxidase 2 (DUOX2) generates H2O2 that plays a critical role in both host defense and chronic inflammation. Previously, we demonstrated that the proinflammatory mediators IFN-γ and LPS enhance expression of DUOX2 and its maturation factor DUOXA2 through STAT1- and NF-κB‒mediated signaling in human pancreatic cancer cells. Using a panel of colon and pancreatic cancer cell lines, we now report the induction of DUOX2/DUOXA2 mRNA and protein expression by the TH2 cytokine IL-4. IL-4 activated STAT6 signaling that, when silenced, significantly decreased induction of DUOX2. Furthermore, the TH17 cytokine IL-17A combined synergistically with IL-4 to increase DUOX2 expression in both colon and pancreatic cancer cells mediated, at least in part, by signaling through NF-κB. The upregulation of DUOX2 was associated with a significant increase in the production of extracellular H2O2 and DNA damage-as indicated by the accumulation of 8-oxo-dG and γH2AX-which was suppressed by the NADPH oxidase inhibitor diphenylene iodonium and a DUOX2-specific small interfering RNA. The clinical relevance of these experiments is suggested by immunohistochemical, microarray, and quantitative RT-PCR studies of human colon and pancreatic tumors demonstrating significantly higher DUOX2, IL-4R, and IL-17RA expression in tumors than in adjacent normal tissues; in pancreatic adenocarcinoma, increased DUOX2 expression is adversely associated with overall patient survival. These data suggest a functional association between DUOX2-mediated H2O2 production and induced DNA damage in gastrointestinal malignancies.


Subject(s)
Colonic Neoplasms/metabolism , DNA Damage , Dual Oxidases/genetics , Hydrogen Peroxide/metabolism , Interleukin-17/pharmacology , Interleukin-4/pharmacology , Pancreatic Neoplasms/metabolism , Cell Line, Tumor , Colonic Neoplasms/pathology , Humans , NF-kappa B/physiology , Oxidation-Reduction , Pancreatic Neoplasms/pathology , Receptors, Interleukin-4/physiology , STAT6 Transcription Factor/physiology , Signal Transduction , Up-Regulation
12.
Oxid Med Cell Longev ; 2018: 4967318, 2018.
Article in English | MEDLINE | ID: mdl-30116483

ABSTRACT

The introduction of intracameral anaesthesia by injection of lidocaine has become popular in cataract surgery for its inherent potency, rapid onset, tissue penetration, and efficiency. However, intracameral lidocaine causes corneal thickening, opacification, and corneal endothelial cell loss. Herein, we investigated the effects of lidocaine combined with sodium ferulate, an antioxidant with antiapoptotic and anti-inflammatory properties, on lidocaine-induced damage of corneal endothelia with in vitro experiment of morphological changes and cell viability of cultured human corneal endothelial cells and in vivo investigation of corneal endothelial cell density and central corneal thickness of cat eyes. Our finding indicates that sodium ferulate from 25 to 200 mg/L significantly reduced 2 g/L lidocaine-induced toxicity to human corneal endothelial cells, and 50 mg/L sodium ferulate recovered the damaged human corneal endothelial cells to normal growth status. Furthermore, 100 mg/L sodium ferulate significantly inhibited lidocaine-induced corneal endothelial cell loss and corneal thickening in cat eyes. In conclusion, sodium ferulate protects human corneal endothelial cells from lidocaine-induced cytotoxicity and attenuates corneal endothelial cell loss and central corneal thickening of cat eyes after intracameral injection with lidocaine. It is likely that the antioxidant effect of sodium ferulate reduces the cytotoxic and inflammatory corneal reaction during intracameral anaesthesia.


Subject(s)
Coumaric Acids/therapeutic use , Endothelium, Corneal/drug effects , Lidocaine/adverse effects , Animals , Cats , Coumaric Acids/pharmacology , Humans , Male
13.
Mol Carcinog ; 56(12): 2643-2662, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28762556

ABSTRACT

NADPH oxidase 5 (NOX5) generated reactive oxygen species (ROS) have been implicated in signaling cascades that regulate cancer cell proliferation. To evaluate and validate NOX5 expression in human tumors, we screened a broad range of tissue microarrays (TMAs), and report substantial overexpression of NOX5 in malignant melanoma and cancers of the prostate, breast, and ovary. In human UACC-257 melanoma cells that possesses high levels of functional endogenous NOX5, overexpression of NOX5 resulted in enhanced cell growth, increased numbers of BrdU positive cells, and increased γ-H2AX levels. Additionally, NOX5-overexpressing (stable and inducible) UACC-257 cells demonstrated increased normoxic HIF-1α expression and decreased p27Kip1 expression. Similarly, increased normoxic HIF-1α expression and decreased p27Kip1 expression were observed in stable NOX5-overexpressing clones of KARPAS 299 human lymphoma cells and in the human prostate cancer cell line, PC-3. Conversely, knockdown of endogenous NOX5 in UACC-257 cells resulted in decreased cell growth, decreased HIF-1α expression, and increased p27Kip1 expression. Likewise, in an additional human melanoma cell line, WM852, and in PC-3 cells, transient knockdown of endogenous NOX5 resulted in increased p27Kip1 and decreased HIF-1α expression. Knockdown of endogenous NOX5 in UACC-257 cells resulted in decreased Akt and GSK3ß phosphorylation, signaling pathways known to modulate p27Kip1 levels. In summary, our findings suggest that NOX5 expression in human UACC-257 melanoma cells could contribute to cell proliferation due, in part, to the generation of high local concentrations of extracellular ROS that modulate multiple pathways that regulate HIF-1α and networks that signal through Akt/GSK3ß/p27Kip1 .


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , NADPH Oxidase 5/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Female , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , NADPH Oxidase 5/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference
14.
Biochem Pharmacol ; 143: 25-38, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28709950

ABSTRACT

The NADPH oxidases (NOXs) play a recognized role in the development and progression of inflammation-associated disorders, as well as cancer. To date, several NOX inhibitors have been developed, through either high throughput screening or targeted disruption of NOX interaction partners, although only a few have reached clinical trials. To improve the efficacy and bioavailability of the iodonium class NOX inhibitor diphenylene iodonium (DPI), we synthesized 36 analogs of DPI, focusing on improved solubility and functionalization. The inhibitory activity of the analogs was interrogated through cell viability and clonogenic studies with a colon cancer cell line (HT-29) that depends on NOX for its proliferative potential. Lack of altered cellular respiration at relevant iodonium analog concentrations was also demonstrated. Additionally, inhibition of ROS generation was evaluated with a luminescence assay for superoxide, or by Amplex Red® assay for H2O2 production, in cell models expressing specific NOX isoforms. DPI and four analogs (NSCs 740104, 751140, 734428, 737392) strongly inhibited HT-29 cell growth and ROS production with nanomolar potency in a concentration-dependent manner. NSC 737392 and 734428, which both feature nitro functional groups at the meta position, had >10-fold higher activity against ROS production by cells that overexpress dual oxidase 2 (DUOX2) than the other compounds examined (IC50≈200-400nM). Based on these results, we synthesized and tested NSC 780521 with optimized potency against DUOX2. Iodonium analogs with anticancer activity, including the first generation of targeted agents with improved specificity against DUOX2, may provide a novel therapeutic approach to NOX-driven tumors.


Subject(s)
Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Onium Compounds/pharmacology , Thiophenes/pharmacology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Dual Oxidases , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , HT29 Cells , Humans , Molecular Structure , NADH, NADPH Oxidoreductases/genetics , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , Onium Compounds/chemical synthesis , Onium Compounds/chemistry , Oxygen Consumption/drug effects , Reactive Oxygen Species/antagonists & inhibitors , Thiophenes/chemical synthesis , Thiophenes/chemistry
15.
Redox Biol ; 13: 182-195, 2017 10.
Article in English | MEDLINE | ID: mdl-28578276

ABSTRACT

NADPH oxidase 4 (NOX4) is a redox active, membrane-associated protein that contributes to genomic instability, redox signaling, and radiation sensitivity in human cancers based on its capacity to generate H2O2 constitutively. Most studies of NOX4 in malignancy have focused on the evaluation of a small number of tumor cell lines and not on human tumor specimens themselves; furthermore, these studies have often employed immunological tools that have not been well characterized. To determine the prevalence of NOX4 expression across a broad range of solid tumors, we developed a novel monoclonal antibody that recognizes a specific extracellular region of the human NOX4 protein, and that does not cross-react with any of the other six members of the NOX gene family. Evaluation of 20 sets of epithelial tumors revealed, for the first time, high levels of NOX4 expression in carcinomas of the head and neck (15/19 patients), esophagus (12/18 patients), bladder (10/19 patients), ovary (6/17 patients), and prostate (7/19 patients), as well as malignant melanoma (7/15 patients) when these tumors were compared to histologically-uninvolved specimens from the same organs. Detection of NOX4 protein upregulation by low levels of TGF-ß1 demonstrated the sensitivity of this new probe; and immunofluorescence experiments found that high levels of endogenous NOX4 expression in ovarian cancer cells were only demonstrable associated with perinuclear membranes. These studies suggest that NOX4 expression is upregulated, compared to normal tissues, in a well-defined, and specific group of human carcinomas, and that its expression is localized on intracellular membranes in a fashion that could modulate oxidative DNA damage.


Subject(s)
Gene Expression Regulation, Neoplastic , NADPH Oxidase 4/metabolism , Carcinoma/genetics , Carcinoma/metabolism , Cell Line, Tumor , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Female , HEK293 Cells , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Male , NADPH Oxidase 4/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Oxidative Stress , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism
16.
Oncotarget ; 8(24): 38113-38135, 2017 Jun 13.
Article in English | MEDLINE | ID: mdl-28498822

ABSTRACT

Human colon cancers express higher levels of NADPH oxidase 1 [NOX1] than adjacent normal epithelium. It has been suggested that reactive oxygen species [ROS] derived from NOX1 contribute to DNA damage and neoplastic transformation in the colon, particularly during chronic inflammatory stress. However, the mechanism(s) underlying increased NOX1 expression in malignant tumors or chronic inflammatory states involving the intestine are poorly characterized. We examined the effects of two pro-inflammatory cytokines, IL-4 and IL-13, on the regulation of NOX1. NOX1 expression was increased 4- to 5-fold in a time- and concentration-dependent manner by both cytokines in human colon cancer cell lines when a functional Type II IL-4 receptor was present. Increased NOX1 transcription following IL-4/IL-13 exposure was mediated by JAK1/STAT6 signaling, was associated with a ROS-related inhibition of protein tyrosine phosphatase activity, and was dependent upon activation and specific binding of GATA3 to the NOX1 promoter. NOX1-mediated ROS production increased cell cycle progression through S-phase leading to a significant increase in cellular proliferation. Evaluation of twenty pairs of surgically-resected colon cancers and their associated uninvolved adjacent colonic epithelium demonstrated a significant increase in the active form of NOX1, NOX1-L, in tumors compared to normal tissues, and a significant correlation between the expression levels of NOX1 and the Type II IL-4 receptor in tumor and the uninvolved colon. These studies imply that NOX1 expression, mediated by IL-4/IL-13, could contribute to an oxidant milieu capable of supporting the initiation or progression of colonic cancer, suggesting a role for NOX1 as a therapeutic target.


Subject(s)
Colonic Neoplasms/metabolism , Interleukin-13/metabolism , Interleukin-4/metabolism , NADPH Oxidase 1/metabolism , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/pathology , Humans , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism
17.
J Biol Chem ; 292(19): 7866-7887, 2017 05 12.
Article in English | MEDLINE | ID: mdl-28330872

ABSTRACT

Reactive oxygen species (ROS) play a critical role in cell signaling and proliferation. NADPH oxidase 1 (NOX1), a membrane-bound flavin dehydrogenase that generates O2̇̄, is highly expressed in colon cancer. To investigate the role that NOX1 plays in colon cancer growth, we used shRNA to decrease NOX1 expression stably in HT-29 human colon cancer cells. The 80-90% decrease in NOX1 expression achieved by RNAi produced a significant decline in ROS production and a G1/S block that translated into a 2-3-fold increase in tumor cell doubling time without increased apoptosis. The block at the G1/S checkpoint was associated with a significant decrease in cyclin D1 expression and profound inhibition of mitogen-activated protein kinase (MAPK) signaling. Decreased steady-state MAPK phosphorylation occurred concomitant with a significant increase in protein phosphatase activity for two colon cancer cell lines in which NOX1 expression was knocked down by RNAi. Diminished NOX1 expression also contributed to decreased growth, blood vessel density, and VEGF and hypoxia-inducible factor 1α (HIF-1α) expression in HT-29 xenografts initiated from NOX1 knockdown cells. Microarray analysis, supplemented by real-time PCR and Western blotting, revealed that the expression of critical regulators of cell proliferation and angiogenesis, including c-MYC, c-MYB, and VEGF, were down-regulated in association with a decline in hypoxic HIF-1α protein expression downstream of silenced NOX1 in both colon cancer cell lines and xenografts. These studies suggest a role for NOX1 in maintaining the proliferative phenotype of some colon cancers and the potential of NOX1 as a therapeutic target in this disease.


Subject(s)
Colonic Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Animals , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Colon/metabolism , Cyclin D1/metabolism , HT29 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , NADPH Oxidase 1 , Neoplasm Transplantation , Phenotype , Phosphorylation , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Vascular Endothelial Growth Factor A/metabolism
18.
Cell Stress Chaperones ; 21(6): 1077-1088, 2016 11.
Article in English | MEDLINE | ID: mdl-27581971

ABSTRACT

Upon diapause termination and exposure to favorable environmental conditions, cysts of the crustacean Artemia franciscana reinitiate development, a process dependent on the resumption of metabolic activity and the maintenance of protein homeostasis. The objective of the work described herein was to characterize molecular chaperones during post-diapause growth of A. franciscana. An Hsp40 complementary DNA (cDNA) termed ArHsp40 was cloned and shown to encode a protein with an amino-terminal J-domain containing a conserved histidine, proline, and aspartic acid (HPD) motif. Following the J-domain was a Gly/Phe (G/F) rich domain, a zinc-binding domain which contained a modified CXXCXGXG motif, and the carboxyl-terminal substrate binding region, all characteristics of type I Hsp40. Multiple alignment and protein modeling showed that ArHsp40 is comparable to Hsp40s from other eukaryotes and likely to be functionally similar. qRT-PCR revealed that during post-diapause development, ArHsp40 messenger RNA (mRNA) varied slightly until the E2/E3 stage and decreased significantly upon hatching. The immunoprobing of Western blots demonstrated that ArHsp40 was also relatively constant until E2/E3 and then declined dramatically. The drop in ArHsp40 when metabolism and protein synthesis were increasing was unexpected and demonstrated developmental regulation. The reduction in ArHsp40 at such an active life history stage indicates, as one possibility, that A. franciscana possesses additional Hsp40s, one or more of which replaces ArHsp40 as development progresses. Increased synthesis upon heat shock established that in addition to being developmentally regulated, ArHsp40 is stress inducible and, because it is found in mature cysts, ArHsp40 has the potential to contribute to stress tolerance during diapause.


Subject(s)
Artemia/metabolism , Arthropod Proteins/metabolism , HSP40 Heat-Shock Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Artemia/growth & development , Arthropod Proteins/genetics , Cloning, Molecular , Diapause , HSP40 Heat-Shock Proteins/genetics , Heat-Shock Response , Larva/metabolism , Molecular Sequence Data , RNA, Messenger/metabolism , Sequence Alignment , Temperature
19.
Oncotarget ; 7(42): 68412-68433, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27637085

ABSTRACT

Several NADPH oxidase family members, including dual oxidase 2 [DUOX2], are expressed in human tumors, particularly gastrointestinal cancers associated with long-standing chronic inflammation. We found previously that exposure of pancreatic ductal adenocarcinoma cells to the pro-inflammatory cytokine IFN-γ increased DUOX2 expression (but not other NADPH oxidases) leading to long-lived H2O2 production. To elucidate the pathophysiology of DUOX2-mediated H2O2 formation in the pancreas further, we demonstrate here that IFN-γ-treated BxPC-3 and CFPAC-1 pancreatic cancer cells (known to increase DUOX2 expression) produce significant levels of intracellular oxidants and extracellular H2O2 which correlate with concomitant up-regulation of VEGF-A and HIF-1α transcription. These changes are not observed in the PANC-1 line that does not increase DUOX2 expression following IFN-γ treatment. DUOX2 knockdown with short interfering RNA significantly decreased IFN-γ-induced VEGF-A or HIF-1α up-regulation, as did treatment of pancreatic cancer cells with the NADPH oxidase inhibitor diphenylene iodonium, the multifunctional reduced thiol N-acetylcysteine, and the polyethylene glycol-modified form of the hydrogen peroxide detoxifying enzyme catalase. Increased DUOX2-related VEGF-A expression appears to result from reactive oxygen-mediated activation of ERK signaling that is responsible for AP-1-related transcriptional effects on the VEGF-A promoter. To clarify the relevance of these observations in vivo, we demonstrate that many human pre-malignant pancreatic intraepithelial neoplasms and frank pancreatic cancers express substantial levels of DUOX protein compared to histologically normal pancreatic tissues, and that expression of both DUOX2 and VEGF-A mRNAs is significantly increased in surgically-resected pancreatic cancers compared to the adjacent normal pancreas.


Subject(s)
Adenocarcinoma/genetics , Dual Oxidases/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Pancreatic Neoplasms/genetics , Vascular Endothelial Growth Factor A/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Dual Oxidases/antagonists & inhibitors , Dual Oxidases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Hydrogen Peroxide/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interferon-gamma/pharmacology , Mice, Nude , Onium Compounds/pharmacology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , RNA Interference , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/metabolism
20.
Clin Sci (Lond) ; 128(12): 863-75, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25818486

ABSTRACT

The mechanism by which reactive oxygen species (ROS) are produced by tumour cells remained incompletely understood until the discovery over the last 15 years of the family of NADPH oxidases (NOXs 1-5 and dual oxidases DUOX1/2) which are structural homologues of gp91phox, the major membrane-bound component of the respiratory burst oxidase of leucocytes. Knowledge of the roles of the NOX isoforms in cancer is rapidly expanding. Recent evidence suggests that both NOX1 and DUOX2 species produce ROS in the gastrointestinal tract as a result of chronic inflammatory stress; cytokine induction (by interferon-γ, tumour necrosis factor α, and interleukins IL-4 and IL-13) of NOX1 and DUOX2 may contribute to the development of colorectal and pancreatic carcinomas in patients with inflammatory bowel disease and chronic pancreatitis, respectively. NOX4 expression is increased in pre-malignant fibrotic states which may lead to carcinomas of the lung and liver. NOX5 is highly expressed in malignant melanomas, prostate cancer and Barrett's oesophagus-associated adenocarcinomas, and in the last it is related to chronic gastro-oesophageal reflux and inflammation. Over-expression of functional NOX proteins in many tissues helps to explain tissue injury and DNA damage from ROS that accompany pre-malignant conditions, as well as elucidating the potential mechanisms of NOX-related damage that contribute to both the initiation and the progression of a wide range of solid and haematopoietic malignancies.


Subject(s)
NADPH Oxidases/metabolism , Neoplasms/enzymology , Hematologic Neoplasms/enzymology , Humans , NADPH Oxidases/genetics , NADPH Oxidases/physiology , Precancerous Conditions/enzymology , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...