Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
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
2.
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
3.
Antioxid Redox Signal ; 33(6): 435-454, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32008376

ABSTRACT

Significance: The primary function of NADPH oxidases (NOX1-5 and dual oxidases DUOX1/2) is to produce reactive oxygen species (ROS). If inadequately regulated, NOX-associated ROS can promote oxidative stress, aberrant signaling, and genomic instability. Correspondingly, NOX isoforms are known to be overexpressed in multiple malignancies, thus constituting potential therapeutic targets in cancer. Recent Advances: Multiple genetic studies aimed at suppressing the expression of NOX proteins in cellular and animal models of cancer have provided support for the notion that NOXs play a pro-tumorigenic role. Further, large drug screens and rational design efforts have yielded inhibitor compounds, such as the diphenylene iodonium (DPI) analog series developed by our group, with increased selectivity and potency over "first generation" NOX inhibitors such as apocynin and DPI. Critical Issues: The precise role of NOX enzymes in tumor biology remains poorly defined. The tumorigenic properties of NOXs vary with cancer type, and precise tools, such as selective inhibitors, are needed to deconvolute NOX contribution to cancer development. Most NOX inhibitors developed to date are unspecific, and/or their mechanistic and pharmacological characteristics are not well defined. A lack of high-resolution crystal structures for NOX functional domains has hindered the development of potent and selective inhibitors. Future Directions: In-depth studies of NOX interactions with the tumor microenvironment (e.g., cytokines, cell-surface antigens) will help identify new approaches for NOX inhibition in cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Neoplasms/metabolism , Animals , Antineoplastic Agents/therapeutic use , Biomarkers , Disease Models, Animal , Enzyme Activation/drug effects , Enzyme Inhibitors/therapeutic use , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/pathology , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Tumor Microenvironment/drug effects
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
Anal Biochem ; 459: 1-11, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24799347

ABSTRACT

Hypoxia-inducible factor-1 alpha (HIF-1α) is an important marker of hypoxia in human tumors and has been implicated in tumor progression. Drugs targeting HIF-1α are being developed, but the ability to measure drug-induced changes in HIF-1α is limited by the lability of the protein in normoxia. Our goal was to devise methods for specimen collection and processing that preserve HIF-1α in solid tumor tissues and to develop and validate a two-site chemiluminescent quantitative enzyme-linked immunosorbent assay (ELISA) for HIF-1α. We tested various strategies for HIF-1α stabilization in solid tumors, including nitrogen gas-purged lysis buffer, the addition of proteasome inhibitors or the prolyl hydroxylase inhibitor 2-hydroxyglutarate, and bead homogenization. Degassing and the addition of 2-hydroxyglutarate to the collection buffer significantly increased HIF-1α recovery, whereas bead homogenization in sealed tubes improved HIF-1α recovery and reduced sample variability. Validation of the ELISA demonstrated intra- and inter-assay variability of less than 15% and accuracy of 99.8±8.3% as assessed by spike recovery. Inter-laboratory reproducibility was also demonstrated (R(2)=0.999). Careful sample handling techniques allow us to quantitatively detect HIF-1α in samples as small as 2.5µg of total protein extract, and this method is currently being applied to analyze tumor biopsy specimens in early-phase clinical trials.


Subject(s)
Enzyme-Linked Immunosorbent Assay , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms/pathology , Specimen Handling/methods , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic , Female , Humans , Mice
13.
Cancer Lett ; 345(2): 164-73, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-23988267

ABSTRACT

Although it is now accepted that chronic inflammation plays an essential role in tumorigenesis, the underlying molecular mechanisms linking inflammation and cancer remain to be fully explored. Inflammatory mediators present in the tumor microenvironment, including cytokines and growth factors, as well as reactive oxygen species (ROS) and reactive nitrogen species (RNS), have been implicated in the etiology of inflammation-associated cancers. Epithelial NADPH oxidase (Nox) family proteins, which generate ROS regulated by cytokines, are upregulated during chronic inflammation and cancer. ROS serve as effector molecules participating in host defense or as chemo-attractants recruiting leukocytes to wounds, thereby influencing the inflammatory reaction in damaged tissues. ROS can alter chromosomal DNA, leading to genomic instability, and may serve as signaling molecules that affect tumor cell proliferation, survival, metabolism, angiogenesis, and metastasis. Targeting Noxs and their downstream signaling components may be a promising approach to pre-empting inflammation-related malignancies.


Subject(s)
Inflammation/complications , Neoplasms/etiology , Animals , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Chronic Disease , Humans , Inflammation/drug therapy , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/metabolism , NADPH Oxidases/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Signal Transduction , Transcription Factors/metabolism , Tumor Microenvironment
14.
Antioxid Redox Signal ; 20(17): 2873-89, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24156355

ABSTRACT

SIGNIFICANCE: Reactive oxygen species (ROS) promote genomic instability, altered signal transduction, and an environment that can sustain tumor formation and growth. The NOX family of NADPH oxidases, membrane-bound epithelial superoxide and hydrogen peroxide producers, plays a critical role in the maintenance of immune function, cell growth, and apoptosis. The impact of NOX enzymes in carcinogenesis is currently being defined and may directly link chronic inflammation and NOX ROS-mediated tumor formation. RECENT ADVANCES: Increased interest in the function of NOX enzymes in tumor biology has spurred a surge of investigative effort to understand the variability of NOX expression levels in tumors and the effect of NOX activity on tumor cell proliferation. These initial efforts have demonstrated a wide variance in NOX distribution and expression levels across numerous cancers as well as in common tumor cell lines, suggesting that much remains to be discovered about the unique role of NOX-related ROS production within each system. Progression from in vitro cell line studies toward in vivo tumor tissue screening and xenograft models has begun to provide evidence supporting the importance of NOX expression in carcinogenesis. CRITICAL ISSUES: A lack of universally available, isoform-specific antibodies and animal tumor models of inducible knockout or over-expression of NOX isoforms has hindered progress toward the completion of in vivo studies. FUTURE DIRECTIONS: In vivo validation experiments and the use of large, existing gene expression data sets should help define the best model systems for studying the NOX homologues in the context of cancer.


Subject(s)
Cell Proliferation/genetics , NADPH Oxidases/metabolism , Neoplasms/genetics , Signal Transduction/genetics , Apoptosis/genetics , Cell Cycle/genetics , Humans , Hydrogen Peroxide/metabolism , NADPH Oxidases/genetics , Neoplasms/physiopathology , Neoplasms/therapy , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Superoxides/metabolism
15.
Free Radic Biol Med ; 65: 497-508, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23851018

ABSTRACT

Reactive oxygen species generated by NADPH oxidase 5 (Nox5) have been implicated in physiological and pathophysiological signaling pathways, including cancer development and progression. However, because immunological tools are lacking, knowledge of the role of Nox5 in tumor biology has been limited; the expression of Nox5 protein across tumors and normal tissues is essentially unknown. Here, we report the characterization and use of a mouse monoclonal antibody against a recombinant Nox5 protein (bp 600-746) for expression profiling of Nox5 in human tumors by tissue microarray analysis. Using our novel antibody, we also report the detection of endogenous Nox5 protein in human UACC-257 melanoma cells. Immunofluorescence, confocal microscopy, and immunohistochemical techniques were employed to demonstrate Nox5 localization throughout UACC-257 cells, with perinuclear enhancement. Tissue microarray analysis revealed, for the first time, substantial Nox5 overexpression in several human cancers, including those of prostate, breast, colon, lung, brain, and ovary, as well as in malignant melanoma and non-Hodgkin lymphoma; expression in most nonmalignant tissues was negative to weak. This validated mouse monoclonal antibody will promote further exploration of the functional significance of Nox5 in human pathophysiology, including tumor cell growth and proliferation.


Subject(s)
Antibodies, Monoclonal , Biomarkers, Tumor/analysis , Membrane Proteins/biosynthesis , NADPH Oxidases/biosynthesis , Neoplasms/enzymology , Animals , Blotting, Western , Cell Line, Tumor , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Male , Membrane Proteins/analysis , Mice , Microscopy, Confocal , NADPH Oxidase 5 , NADPH Oxidases/analysis , Real-Time Polymerase Chain Reaction , Tissue Array Analysis
16.
Int J Oncol ; 42(4): 1229-38, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23404210

ABSTRACT

Dual oxidase 2 (Duox2), one of the seven members of the NADPH oxidase gene family, plays a critical role in generating H2O2 for thyroid hormone biosynthesis and as an integral part of the host defense system of the respiratory epithelium and the gastrointestinal tract. Recent evidence suggests that the regulation of Duox2 expression is under the control of pro-inflammatory cytokines and that Duox2-induced reactive oxygen species (ROS) contribute to the inflammation-related tissue injury that occurs in two pre-malignant, inflammatory conditions: chronic pancreatitis and inflammatory bowel disease. Because no reliable Duox antibodies are commercially available, we report the development of a murine monoclonal antibody (MAb) to Duox2 (clone Duox S-12) and its use for the characterization of Duox2 expression in human tumors, tumor cell lines and normal tissues. Duox S-12 specifically detected both endogenously- and ectopically-expressed Duox2 protein by immunoblotting, immunofluorescence microscopy and immunohistochemistry (where both membranous and cytoplasmic staining were present). Duox2 expression detected by Duox S-12 was functionally coupled to the generation of H(2)O(2) in pancreatic cancer cells that expressed Duox2 and its cognate maturation factor DuoxA2. Although Duox S-12 recognizes ectopically expressed Duox1 protein because of the extensive amino acid homology between Duox1 and Duox2, the lack of substantial Duox1 mRNA expression in human tumors (except thyroid cancer) allowed us to evaluate Duox2 expression across a wide range of normal and malignant tissues by immuno-histochemistry. Duox2 was expressed at elevated levels in many human cancers, most notably tumors of the prostate, lung, colon and breast while brain tumors and lymphomas demonstrated the lowest frequency of expression. The Duox-specific monoclonal antibody described here provides a promising tool for the further examination of the role of Duox-dependent reactive oxygen production in inflammation-related carcinogenesis, where alterations in oxidant tone play a critical role in cell growth and proliferation.


Subject(s)
Adenocarcinoma/enzymology , Antibodies, Monoclonal, Murine-Derived/immunology , Breast Neoplasms/enzymology , NADPH Oxidases/metabolism , Pancreatic Neoplasms/enzymology , Amino Acid Sequence , Animals , Antibodies, Monoclonal, Murine-Derived/chemistry , Antibody Specificity , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Dual Oxidases , Female , Humans , Hybridomas , Hydrogen Peroxide/metabolism , Immunohistochemistry , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , NADPH Oxidases/immunology , Tissue Array Analysis
17.
J Immunol ; 190(4): 1859-72, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23296709

ABSTRACT

Pancreatitis is associated with release of proinflammatory cytokines and reactive oxygen species and plays an important role in the development of pancreatic cancer. We recently demonstrated that dual oxidase (Duox)2, an NADPH oxidase essential for reactive oxygen species-related, gastrointestinal host defense, is regulated by IFN-γ-mediated Stat1 binding to the Duox2 promoter in pancreatic tumor lines. Because LPS enhances the development and invasiveness of pancreatic cancer in vivo following TLR4-related activation of NF-κB, we examined whether LPS, alone or combined with IFN-γ, regulated Duox2. We found that upregulation of TLR4 by IFN-γ in BxPC-3 and CFPAC-1 pancreatic cancer cells was augmented by LPS, resulting in activation of NF-κB, accumulation of NF-κB (p65) in the nucleus, and increased binding of p65 to the Duox2 promoter. TLR4 silencing with small interfering RNAs, as well as two independent NF-κB inhibitors, attenuated LPS- and IFN-γ-mediated Duox2 upregulation in BxPC-3 cells. Induction of Duox2 expression by IFN-γ and LPS may result from IFN-γ-related activation of Stat1 acting in concert with NF-κB-related upregulation of Duox2. Sustained extracellular accumulation of H(2)O(2) generated by exposure to both LPS and IFN-γ was responsible for an ∼50% decrease in BxPC-3 cell proliferation associated with a G(1) cell cycle block, apoptosis, and DNA damage. We also demonstrated upregulation of Duox expression in vivo in pancreatic cancer xenografts and in patients with chronic pancreatitis. These results suggest that inflammatory cytokines can interact to produce a Duox-dependent pro-oxidant milieu that could increase the pathologic potential of pancreatic inflammation and pancreatic cancer cells.


Subject(s)
Interferon-gamma/physiology , Lipopolysaccharides/physiology , Membrane Proteins/biosynthesis , NADPH Oxidases/biosynthesis , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cell Line, Tumor , Chronic Disease , Dual Oxidases , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/enzymology , Pancreatitis/enzymology , Pancreatitis/immunology , Pancreatitis/metabolism , Random Allocation , Reactive Oxygen Species/metabolism , Signal Transduction/immunology , Toll-Like Receptor 4/physiology , Tumor Cells, Cultured
18.
Biochem Pharmacol ; 83(9): 1195-207, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22305747

ABSTRACT

Flavoprotein-dependent reactive oxygen species (ROS) play a critical role in cytokine-mediated signal transduction in normal tissues and tumor cells. The flavoenzyme inhibitors diphenylene iodonium (DPI) and di-2-thienyliodonium (DTI) have been used to inhibit membrane-bound, flavoprotein-containing NADPH oxidases, including epithelial and leukocyte NADPH oxidases (Nox1-5 and Duox 1 and 2). Recent evidence suggests that DPI can decrease tumor cell proliferation; however, the molecular mechanisms involved remain poorly defined. To explore the mechanisms underlying DPI- and DTI-related tumor cell growth delay, we examined growth inhibition patterns produced by both agents in the NCI-60 tumor panel, and determined expression levels of Nox gene family members across these cell lines. Possible molecular targets were predicted using the COMPARE program. DPI was more potent than DTI (GI(50): 10nM versus 10µM); DPI and DTI exposure produced unique patterns of growth inhibition when evaluated against the small molecule anticancer database of the National Cancer Institute. Growth inhibition profiling of DPI revealed a modest positive correlation with Nox1 levels; novel mechanisms of DPI and DTI action, including alterations in Stat, Erk1/2, and Akt pathways, were inferred by correlation with NCI-60 Affymetrix(®) array data. Exposure of HT-29 colon cancer cells, which express Nox1, to DPI and DTI confirmed their inhibitory effects on steady state ROS levels, and demonstrated decreased Stat, Erk1/2, and Akt signaling mediated by IL-4, IL-6, IL-13, and IL-22, possibly due to a concomitant increase in tumor cell phosphatase activity. These findings suggest that DPI and DTI may act therapeutically by altering ROS-related signal transduction.


Subject(s)
Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , NADPH Oxidases/genetics , Onium Compounds/pharmacology , Thiophenes/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cytokines/metabolism , Drug Screening Assays, Antitumor , Gene Expression Regulation/drug effects , HT29 Cells , Humans , Mitochondria/drug effects , Mitochondria/metabolism , NADPH Oxidase 1 , NADPH Oxidases/metabolism , Oligonucleotide Array Sequence Analysis , Phosphoprotein Phosphatases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
19.
J Med Chem ; 54(14): 4937-53, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21710981

ABSTRACT

The DNA-relaxing enzyme topoisomerase I (Top1) can be inhibited by heterocyclic compounds such as indolocarbazoles and indenoisoquinolines. Carbohydrate and hydroxyl-containing side chains are essential for the biological activity of indolocarbazoles. The current study investigated how similar functionalities could be "translated" to the indenoisoquinoline system and how stereochemistry and hydrogen bonding affect biological activity. Herein is described the preparation and assay of indenoisoquinolines substituted with short-chain alcohols, diols, and carbohydrates. Several compounds (including those derived from sugars) display potent Top1 poisoning and antiproliferative activities. The Top1 poisoning activity of diol-substituted indenoisoquinolines is dependent upon stereochemistry. Although the effect is striking, molecular modeling and docking studies do not indicate any reason for the difference in activity due to similar calculated interactions between the ligand and Top1-DNA complex and ambiguity about the binding mode. A stereochemical dependence was also observed for carbohydrate-derived indenoisoquinolines. Although similar trends were observed in other classes of Top1 inhibitors, the exact nature of this effect has yet to be elucidated.


Subject(s)
Alcohols/chemical synthesis , Hexoses/chemical synthesis , Indenes/chemical synthesis , Pentoses/chemical synthesis , Quinolines/chemical synthesis , Topoisomerase I Inhibitors/chemical synthesis , Alcohols/chemistry , Alcohols/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Hexoses/chemistry , Hexoses/pharmacology , Humans , Hydrogen Bonding , Indenes/chemistry , Indenes/pharmacology , Models, Molecular , Pentoses/chemistry , Pentoses/pharmacology , Quinolines/chemistry , Quinolines/pharmacology , Stereoisomerism , Structure-Activity Relationship , Topoisomerase I Inhibitors/chemistry , Topoisomerase I Inhibitors/pharmacology
20.
J Biol Chem ; 286(14): 12245-56, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21321110

ABSTRACT

Dual oxidase 2 is a member of the NADPH oxidase (Nox) gene family that plays a critical role in the biosynthesis of thyroid hormone as well as in the inflammatory response of the upper airway mucosa and in wound healing, presumably through its ability to generate reactive oxygen species, including H2O2. The recently discovered overexpression of Duox2 in gastrointestinal malignancies, as well as our limited understanding of the regulation of Duox2 expression, led us to examine the effect of cytokines and growth factors on Duox2 in human tumor cells. We found that exposure of human pancreatic cancer cells to IFN-γ (but not other agents) produced a profound up-regulation of the expression of Duox2, and its cognate maturation factor DuoxA2, but not other members of the Nox family. Furthermore, increased Duox2/DuoxA2 expression was closely associated with a significant increase in the production of both intracellular reactive oxygen species and extracellular H2O2. Examination of IFN-γ-mediated signaling events demonstrated that in addition to the canonical Jak-Stat1 pathway, IFN-γ activated the p38-MAPK pathway in pancreatic cancer cells, and both played an important role in the induction of Duox2 by IFN-γ. Duox2 up-regulation following IFN-γ exposure is also directly associated with the binding of Stat1 to elements of the Duox2 promoter. Our findings suggest that the pro-inflammatory cytokine IFN-γ initiates a Duox2-mediated reactive oxygen cascade in human pancreatic cancer cells; reactive oxygen species production in this setting could contribute to the pathophysiologic characteristics of these tumors.


Subject(s)
Interferon-gamma/pharmacology , Membrane Proteins/metabolism , NADPH Oxidases/metabolism , STAT1 Transcription Factor/metabolism , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Dual Oxidases , Flow Cytometry , Humans , Hydrogen Peroxide/metabolism , Membrane Proteins/genetics , Microscopy, Confocal , NADPH Oxidases/genetics , Onium Compounds/pharmacology , Polymerase Chain Reaction , Protein Binding/drug effects , Reactive Oxygen Species/metabolism , STAT1 Transcription Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...