Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Biol Chem ; 290(45): 27090-27100, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26385919

ABSTRACT

Arsenic (As(3+)) is a carcinogen with considerable environmental and occupational relevancy. The present study shows that As(3+)-transformed human lung bronchial epithelial BEAS-2B cells (AsT cells) exhibit the property of apoptosis resistance. The level of basal reactive oxygen species (ROS) is very low in AsT cells in correlation with elevated expressions of both antioxidant enzymes and antiapoptotic proteins. Nuclear factor erythroid 2-related factor (Nrf2) and p62 are constitutively expressed. These two proteins up-regulate antioxidant enzymes and antiapoptotic proteins. The knockdown of Nrf2 or p62 by small interfering RNA (siRNA) enhanced both ROS levels and As(3+)-induced apoptosis in transformed cells. AsT cells have autophagy deficiency as evidenced by reduced formation of microtubule-associated protein 1 light chain 3 (LC3)-II, GFP-LC3 puncta, and autophagy flux. Results obtained using a soft agar assay and shRNA Nrf2-transfected cells show that Nrf2 plays an antioncogenic role before transformation, whereas this transcription factor plays an oncogenic role after transformation. In addition, depletion of Nrf2 by shRNA dramatically inhibited growth and proliferation of transformed cells. Furthermore, the Nrf2 protein levels and antiapoptotic and antioxidant enzyme levels are higher in lung adenocarcinoma than in normal tissues. Collectively, this study demonstrates that a constitutively high level of Nrf2 in AsT cells up-regulates the antioxidant proteins catalase and superoxide dismutase as well as the antiapoptotic proteins Bcl-2 and Bcl-xL. The final consequences are decreased ROS generation and increased apoptotic resistance, cell survival and proliferation, and tumorigenesis.


Subject(s)
Arsenic/toxicity , Carcinogenesis/chemically induced , Carcinogenesis/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Antioxidants/metabolism , Apoptosis , Autophagy , Carcinogenesis/metabolism , Catalase/metabolism , Cell Line , Cell Survival , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Gene Knockdown Techniques , Genes, Tumor Suppressor , Humans , Models, Biological , NF-E2-Related Factor 2/antagonists & inhibitors , Oncogenes , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , bcl-X Protein/genetics , bcl-X Protein/metabolism
2.
Toxicol Appl Pharmacol ; 307: 81-90, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27470422

ABSTRACT

Hexavalent chromium (Cr(VI)) is classified as a human carcinogen. Cr(VI) has been associated with adenocarcinomas and squamous cell carcinoma of the lung. The present study shows that acute Cr(VI) treatment in human bronchial epithelial cells (BEAS-2B) increased inflammatory responses (TNF-α, COX-2, and NF-кB/p65) and expression of Nrf2. Cr(VI)-induced generation of reactive oxygen species (ROS) are responsible for increased inflammation. Despite the fact that Nrf2 is a master regulator of response to oxidative stress, silencing of Nrf2 in the acute Cr(VI) treatment had no effect on Cr(VI)-induced inflammation. In contrast, in Cr(VI)-transformed (CrT) cells, Nrf2 is constitutively activated. Knock-down of this protein resulted in decreased inflammation, while silencing of SOD2 and CAT had no effect in the expression of these inflammatory proteins. Results obtained from the knock-down of Nrf2 in CrT cells are very different from the results obtained in the acute Cr(VI) treatment. In BEAS-2B cells, knock-down of Nrf2 had no effect in the inflammation levels, while in CrT cells a decrease in the expression of inflammation markers was observed. These results indicate that before transformation, ROS plays a critical role while Nrf2 not in Cr(VI)-induced inflammation, whereas after transformation (CrT cells), Nrf2 is constitutively activated and this protein maintains inflammation while ROS not. Constitutively high levels of Nrf2 in CrT binds to the promoter regions of COX-2 and TNF-α, leading to increased inflammation. Collectively, our results demonstrate that before cell transformation ROS are important in Cr(VI)-induced inflammation and after transformation a constitutively high level of Nrf2 is important.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Chromium/toxicity , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Cell Line , Cyclooxygenase 2/metabolism , Humans , NF-E2-Related Factor 2/genetics , Oxidative Stress/drug effects , Tumor Necrosis Factor-alpha/metabolism
3.
J Biol Chem ; 289(41): 28660-75, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25157103

ABSTRACT

The cadmium-transformed human lung bronchial epithelial BEAS-2B cells exhibit a property of apoptosis resistance as compared with normal non-transformed BEAS-2B cells. The level of basal reactive oxygen species (ROS) is extremely low in transformed cells in correlation with elevated expressions of both antioxidant enzymes (catalase, SOD1, and SOD2) and antiapoptotic proteins (Bcl-2/Bcl-xL). Moreover, Nrf2 and p62 are highly expressed in these transformed cells. The knockdown of Nrf2 or p62 by siRNA enhances ROS levels and cadmium-induced apoptosis. The binding activities of Nrf2 on the antioxidant response element promoter regions of p62/Bcl-2/Bcl-xL were dramatically increased in the cadmium-exposed transformed cells. Cadmium exposure increased the formation of LC3-II and the frequency of GFP-LC3 punctal cells in non-transformed BEAS-2B cells, whereas these increases are not shown in transformed cells, an indication of autophagy deficiency of transformed cells. Furthermore, the expression levels of Nrf2 and p62 are dramatically increased during chronic long term exposure to cadmium in the BEAS-2B cells as well as antiapoptotic proteins and antioxidant enzymes. These proteins are overexpressed in the tumor tissues derived from xenograft mouse models. Moreover, the colony growth is significantly attenuated in the transformed cells by siRNA transfection specific for Nrf2 or p62. Taken together, this study demonstrates that cadmium-transformed cells have acquired autophagy deficiency, leading to constitutive p62 and Nrf2 overexpression. These overexpressions up-regulate the antioxidant proteins catalase and SOD and the antiapoptotic proteins Bcl-2 and Bcl-xL. The final consequences are decrease in ROS generation, apoptotic resistance, and increased cell survival, proliferation, and tumorigenesis.


Subject(s)
Cadmium/toxicity , Cell Transformation, Neoplastic/genetics , Epithelial Cells/drug effects , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , NF-E2-Related Factor 2/genetics , RNA-Binding Proteins/genetics , Animals , Apoptosis/genetics , Autophagy/genetics , Bronchi/drug effects , Bronchi/metabolism , Bronchi/pathology , Catalase/genetics , Catalase/metabolism , Cell Line , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Neoplasm Transplantation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , bcl-X Protein/genetics , bcl-X Protein/metabolism
4.
Toxicol Appl Pharmacol ; 288(2): 232-9, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26220687

ABSTRACT

Arsenic is a known carcinogen to humans, and chronic exposure to environmental arsenic is a worldwide health concern. As a dietary factor, ethanol carries a well-established risk for malignancies, but the effects of co-exposure to arsenic and ethanol on tumor development are not well understood. In the present study, we hypothesized that ethanol would enhance the function of an environmental carcinogen such as arsenic through increase in COX-2 expression. Our in vitro results show that ethanol enhanced arsenic-induced COX-2 expression. We also show that the increased COX-2 expression associates with intracellular ROS generation, up-regulated AKT signaling, with activation of both NFAT and NF-κB pathways. We demonstrate that antioxidant enzymes have an inhibitory effect on arsenic/ethanol-induced COX-2 expression, indicating that the responsive signaling pathways from co-exposure to arsenic and ethanol relate to ROS generation. In vivo results also show that co-exposure to arsenic and ethanol increased COX-2 expression in mice. We conclude that ethanol enhances arsenic-induced COX-2 expression in colorectal cancer cells via both the NFAT and NF-κB pathways. These results imply that, as a common dietary factor, ethanol ingestion may be a compounding risk factor for arsenic-induced carcinogenesis/cancer development.


Subject(s)
Alcohol Drinking/metabolism , Arsenites/toxicity , Carcinogens, Environmental/toxicity , Colorectal Neoplasms/enzymology , Cyclooxygenase 2/biosynthesis , Ethanol/toxicity , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Signal Transduction/drug effects , Sodium Compounds/toxicity , Alcohol Drinking/genetics , Colorectal Neoplasms/genetics , Cyclooxygenase 2/genetics , Dose-Response Relationship, Drug , Enzyme Induction , HCT116 Cells , HT29 Cells , Humans , Male , NF-kappa B/genetics , NFATC Transcription Factors/genetics , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Risk Assessment
5.
Toxicol Appl Pharmacol ; 284(1): 92-99, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25680589

ABSTRACT

Extensive exposure of solar ultraviolet-B (UVB) radiation to skin induces oxidative stress and inflammation that play a crucial role in the induction of skin cancer. Photochemoprevention with natural products represents a simple but very effective strategy for the management of cutaneous neoplasia. In this study, we investigated whether blackberry extract (BBE) reduces chronic inflammatory responses induced by UVB irradiation in SKH-1 hairless mice skin. Mice were exposed to UVB radiation (100 mJ/cm(2)) on alternate days for 10 weeks, and BBE (10% and 20%) was applied topically a day before UVB exposure. Our results show that BBE suppressed UVB-induced hyperplasia and reduced infiltration of inflammatory cells in the SKH-1 hairless mice skin. BBE treatment reduced glutathione (GSH) depletion, lipid peroxidation (LPO), and myeloperoxidase (MPO) in mouse skin by chronic UVB exposure. BBE significantly decreased the level of pro-inflammatory cytokines IL-6 and TNF-α in UVB-exposed skin. Likewise, UVB-induced inflammatory responses were diminished by BBE as observed by a remarkable reduction in the levels of phosphorylated MAP Kinases, Erk1/2, p38, JNK1/2 and MKK4. Furthermore, BBE also reduced inflammatory mediators such as cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), and inducible nitric oxide synthase (iNOS) levels in UVB-exposed skin. Treatment with BBE inhibited UVB-induced nuclear translocation of NF-κB and degradation of IκBα in mouse skin. Immunohistochemistry analysis revealed that topical application of BBE inhibited the expression of 8-oxo-7, 8-dihydro-2'-deoxyguanosine (8-oxodG), cyclobutane pyrimidine dimers (CPD), proliferating cell nuclear antigen (PCNA), and cyclin D1 in UVB-exposed skin. Collectively, these data indicate that BBE protects from UVB-induced oxidative damage and inflammation by modulating MAP kinase and NF-κB signaling pathways.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Rubus , Signal Transduction/drug effects , Skin/drug effects , Sunburn/prevention & control , Sunscreening Agents/pharmacology , Ultraviolet Rays , Active Transport, Cell Nucleus , Animals , Anti-Inflammatory Agents/isolation & purification , Antioxidants/isolation & purification , Biomarkers/metabolism , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , DNA Damage , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Fruit , Inflammation Mediators/metabolism , Lipid Peroxidation/drug effects , Mice, Hairless , Neoplasms, Radiation-Induced/enzymology , Neoplasms, Radiation-Induced/immunology , Neoplasms, Radiation-Induced/prevention & control , Phosphorylation , Phytotherapy , Plant Extracts/isolation & purification , Plants, Medicinal , Rubus/chemistry , Skin/enzymology , Skin/immunology , Skin/pathology , Skin Neoplasms/enzymology , Skin Neoplasms/immunology , Skin Neoplasms/prevention & control , Sunburn/enzymology , Sunburn/immunology , Sunburn/pathology , Sunscreening Agents/isolation & purification , Time Factors
8.
Toxicol Appl Pharmacol ; 280(1): 127-37, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25062774

ABSTRACT

Skin cancer is one of the most commonly diagnosed cancers in the United States. Exposure to ultraviolet-B (UVB) radiation induces inflammation and photocarcinogenesis in mammalian skin. Cyanidin-3-glucoside (C3G), a member of the anthocyanin family, is present in various vegetables and fruits especially in edible berries, and displays potent antioxidant and anticarcinogenic properties. In this study, we have assessed the in vivo effects of C3G on UVB irradiation induced chronic inflammatory responses in SKH-1 hairless mice, a well-established model for UVB-induced skin carcinogenesis. Here, we show that C3G inhibited UVB-induced skin damage and inflammation in SKH-1 hairless mice. Our results indicate that C3G inhibited glutathione depletion, lipid peroxidation and myeloperoxidation in mouse skin by chronic UVB exposure. C3G significantly decreased the production of UVB-induced pro-inflammatory cytokines, such as IL-6 and TNF-α, associated with cutaneous inflammation. Likewise, UVB-induced inflammatory responses were diminished by C3G as observed by a remarkable reduction in the levels of phosphorylated MAP kinases, Erk1/2, p38, JNK1/2 and MKK4. Furthermore, C3G also decreased UVB-induced cyclooxygenase-2 (COX-2), PGE2 and iNOS levels, which are well-known key mediators of inflammation and cancer. Treatment with C3G inhibited UVB-induced nuclear translocation of NF-κB and degradation of IκBα in mice skin. Immunofluorescence assay revealed that topical application of C3G inhibited the expression of 8-hydroxy-2'-deoxyguanosine, proliferating cell nuclear antigen, and cyclin D1 in chronic UVB exposed mouse skin. Collectively, these data indicates that C3G can provide substantial protection against the adverse effects of UVB radiation by modulating UVB-induced MAP kinase and NF-κB signaling pathways.


Subject(s)
Anthocyanins/pharmacology , Glucosides/pharmacology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Ultraviolet Rays/adverse effects , Animals , Dose-Response Relationship, Drug , Female , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Mice , Mice, Hairless , Mitogen-Activated Protein Kinases/physiology , NF-kappa B/physiology , Oxidative Stress/physiology , Random Allocation , Signal Transduction/drug effects , Signal Transduction/physiology , Signal Transduction/radiation effects , Skin Absorption/drug effects , Skin Absorption/physiology , Skin Absorption/radiation effects
9.
Toxicol Appl Pharmacol ; 281(2): 230-41, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25448439

ABSTRACT

Hexavalent chromium [Cr(VI)] is a well-known human carcinogen associated with the incidence of lung cancer. Inhibition of metal induced carcinogenesis by a dietary antioxidant is a novel approach. Luteolin, a natural dietary flavonoid found in fruits and vegetables, possesses potent antioxidant and anti-inflammatory activity. We found that short term exposure of human bronchial epithelial cells (BEAS-2B) to Cr(VI) (5µM) showed a drastic increase in ROS generation, NADPH oxidase (NOX) activation, lipid peroxidation, and glutathione depletion, which were significantly inhibited by the treatment with luteolin in a dose dependent manner. Treatment with luteolin decreased AP-1, HIF-1α, COX-2, and iNOS promoter activity induced by Cr(VI) in BEAS-2B cells. In addition, luteolin protected BEAS-2B cells from malignant transformation induced by chronic Cr(VI) exposure. Moreover, luteolin also inhibited the production of pro-inflammatory cytokines (IL-1ß, IL-6, IL-8, TNF-α) and VEGF in chronic Cr(VI) exposed BEAS-2B cells. Western blot analysis showed that luteolin inhibited multiple gene products linked to survival (Akt, Fak, Bcl-2, Bcl-xL), inflammation (MAPK, NF-κB, COX-2, STAT-3, iNOS, TNF-α) and angiogenesis (HIF-1α, VEGF, MMP-9) in chronic Cr(VI) exposed BEAS-2B cells. Nude mice injected with BEAS-2B cells chronically exposed to Cr(VI) in the presence of luteolin showed reduced tumor incidence compared to Cr(VI) alone treated group. Overexpression of catalase (CAT) or SOD2, eliminated Cr(VI)-induced malignant transformation. Overall, our results indicate that luteolin protects BEAS-2B cells from Cr(VI)-induced carcinogenesis by scavenging ROS and modulating multiple cell signaling mechanisms that are linked to ROS. Luteolin, therefore, serves as a potential chemopreventive agent against Cr(VI)-induced carcinogenesis.


Subject(s)
Anticarcinogenic Agents/pharmacology , Bronchi/drug effects , Cell Transformation, Neoplastic/drug effects , Chromium/toxicity , Epithelial Cells/drug effects , Free Radical Scavengers/pharmacology , Lung Neoplasms/prevention & control , Luteolin/pharmacology , Oxidative Stress/drug effects , Potassium Dichromate/toxicity , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Angiogenic Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Bronchi/metabolism , Bronchi/pathology , Cell Line , Cell Survival/drug effects , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cytoprotection , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Inflammation Mediators/metabolism , Lung Neoplasms/chemically induced , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Nude , Transfection , Xenograft Model Antitumor Assays
10.
Toxicol Appl Pharmacol ; 272(1): 108-16, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23743303

ABSTRACT

Environmental exposure to arsenic is known to cause various cancers. There are some potential relationships between cell malignant transformation and C-X-C chemokine receptor type 4 (CXCR4) expressions. Metastasis, one of the major characteristics of malignantly transformed cells, contributes to the high mortality of cells. CXCR4 and its natural chemokine ligand C-X-C motif ligand 12 (CXCL12) play a critical role in metastasis. Therefore, identification of nutritional factors which are able to inhibit CXCR4 is important for protection from environmental arsenic-induced carcinogenesis and for abolishing metastasis of malignantly transformed cells. The present study demonstrates that apigenin (4',5,7-trihydroxyflavone), a natural dietary flavonoid, suppressed CXCR4 expression in arsenic-transformed Beas-2B cells (B-AsT) and several other types of transformed/cancer cells in a dose- and time-dependent manner. Neither proteasome nor lysosome inhibitor had any effect in reducing the apigenin-induced down-regulation of CXCR4, indicating that apigenin-induced down-regulation of CXCR4 is not due to proteolytic degradation. The down-regulation of CXCR4 is mainly due to the inhibition of nuclear factor κB (NF-κB) transcriptional activity. Apigenin also abolished migration and invasion of transformed cells induced by CXCL12. In a xenograft mouse model, apigenin down-regulated CXCR4 expression and suppressed tumor growth. Taken together, our results show that apigenin is a novel inhibitor of CXCR4 expression. This dietary flavonoid has the potential to suppress migration and invasion of transformed cells and prevent environmental arsenic-induced carcinogenesis.


Subject(s)
Apigenin/pharmacology , Cell Movement/drug effects , Neoplasm Invasiveness/pathology , Receptors, CXCR4/biosynthesis , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Cell Transformation, Neoplastic/drug effects , Down-Regulation/drug effects , Humans , Immunoprecipitation , Luciferases/metabolism , Microscopy, Fluorescence , NF-kappa B/antagonists & inhibitors , Plasmids/genetics , Receptors, CXCR4/drug effects , Transfection , Wound Healing/drug effects , Xenograft Model Antitumor Assays
11.
Toxicol Appl Pharmacol ; 255(3): 287-96, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21767558

ABSTRACT

Cadmium is a toxic heavy metal which is environmentally and occupationally relevant. The mechanisms underlying cadmium-induced autophagy are not yet completely understood. The present study shows that cadmium induces autophagy, as demonstrated by the increase of LC3-II formation and the GFP-LC3 puncta cells. The induction of autophagosomes was directly visualized by electron microscopy in cadmium-exposed skin epidermal cells. Blockage of LKB1 or AMPK by siRNA transfection suppressed cadmium-induced autophagy. Cadmium-induced autophagy was inhibited in dominant-negative AMPK-transfected cells, whereas it was accelerated in cells transfected with the constitutively active form of AMPK. mTOR signaling, a negative regulator of autophagy, was downregulated in cadmium-exposed cells. In addition, cadmium generated reactive oxygen species (ROS) at relatively low levels, and caused poly(ADP-ribose) polymerase-1 (PARP) activation and ATP depletion. Inhibition of PARP by pharmacological inhibitors or its siRNA transfection suppressed ATP reduction and autophagy in cadmium-exposed cells. Furthermore, cadmium-induced autophagy signaling was attenuated by either exogenous addition of catalase and superoxide dismutase, or by overexpression of these enzymes. Consequently, these results suggest that cadmium-mediated ROS generation causes PARP activation and energy depletion, and eventually induces autophagy through the activation of LKB1-AMPK signaling and the down-regulation of mTOR in skin epidermal cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy/drug effects , Cadmium/toxicity , Epidermis/drug effects , Protein Serine-Threonine Kinases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Animals , Autophagy/physiology , Cell Line , Epidermis/metabolism , Mice
12.
J Nat Prod ; 71(9): 1569-73, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18666798

ABSTRACT

Two new anticancer antibiotics of the angucycline class, moromycins A and B (1, 2), along with the known microbial metabolites saquayamycin B (3) and fridamycin D (4) were isolated from the ethyl acetate extract of a culture broth of the terrestrial Streptomyces sp. KY002. The structures consist of a tetrangomycin core and various C- and O-glycosidically linked deoxysugars. The chemical structures of the new secondary metabolites were elucidated by 1D and 2D NMR and by mass spectrometry. Moromycin B (2) showed significant cytotoxicity against H-460 human lung cancer and MCF-7 human breast cancer cells.


Subject(s)
Anthraquinones/isolation & purification , Anthraquinones/pharmacology , Antibiotics, Antineoplastic/isolation & purification , Antibiotics, Antineoplastic/pharmacology , Streptomyces/chemistry , Anthraquinones/chemistry , Antibiotics, Antineoplastic/chemistry , Drug Screening Assays, Antitumor , Female , Humans , Kentucky , Microbial Sensitivity Tests , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular
13.
Oncotarget ; 8(32): 52118-52131, 2017 Aug 08.
Article in English | MEDLINE | ID: mdl-28881718

ABSTRACT

Hexavalent chromium [Cr(VI)] is an important human carcinogen associated with pulmonary diseases and lung cancer. Inhibition of Cr(VI)-induced carcinogenesis by a dietary antioxidant is a novel approach. Quercetin is one of the most abundant dietary flavonoids widely present in many fruits and vegetables, possesses potent antioxidant and anticancer properties. MicroRNA-21 (miR-21) is a key oncomiR significantly elevated in the majority of human cancers that exerts its oncogenic activity by targeting the tumor suppressor gene programmed cell death 4 (PDCD4). The present study examined the effect of quercetin on the inhibition of Cr(VI)-induced malignant cell transformation and the role of miR-21-PDCD4 signaling involved. Our results showed that quercetin decreased ROS generation induced by Cr(VI) exposure in BEAS-2B cells. Chronic Cr(VI) exposure induced malignant cell transformation, increased miR-21 expression and caused inhibition of PDCD4, which were significantly inhibited by the treatment of quercetin in a dose dependent manner. Nude mice injected with BEAS-2B cells chronically exposed to Cr(VI) in the presence of quercetin showed reduced tumor incidence compared to Cr(VI) alone treated group. Stable knockdown of miR-21 and overexpression of PDCD4 or catalase in BEAS-2B cells suppressed Cr(VI)-induced malignant transformation and tumorigenesis. Taken together, these results demonstrate that quercetin is able to protect BEAS-2B cells from Cr(VI)-induced carcinogenesis by targeting miR-21-PDCD4 signaling.

14.
Toxicol Sci ; 151(2): 376-87, 2016 06.
Article in English | MEDLINE | ID: mdl-26962057

ABSTRACT

Nickel compounds are known as human carcinogens. Chronic environmental exposure to nickel is a worldwide health concern. Although the mechanisms of nickel-induced carcinogenesis are not well understood, recent studies suggest that stem cells/cancer stem cells are likely important targets. This study examines the role of cancer stem cells in nickel-induced cell transformation. The nontransformed human bronchial epithelial cell line (Beas-2B) was chronically exposed to nickel chloride for 12 months to induce cell transformation. Nickel induced Beas-2B cell transformation, and cancer stem-like cells were enriched in nickel-transformed cell (BNiT) population. The BNiT cancer stem-like cells demonstrated enhanced self-renewal and distinctive differentiation properties. In vivo tumorigenesis studies show that BNiT cancer stem-like cells possess a high tumor-initiating capability. It was also demonstrated that superoxide dismutase 1 was involved in the accumulation of cancer stem-like cells; the regulation of superoxide dismutase 1 expression was different in transformed stem-like cells and nontransformed. Overall, the accumulation of stem-like cells and their enhanced stemness functions contribute to nickel-induced tumorigenesis. Our study provides additional insight into the mechanisms by which metals or other chemicals can induce carcinogenesis.


Subject(s)
Bronchi/drug effects , Cell Transformation, Neoplastic/chemically induced , Epithelial Cells/drug effects , Lung Neoplasms/chemically induced , Neoplastic Stem Cells/drug effects , Nickel/toxicity , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bronchi/metabolism , Bronchi/pathology , Cell Differentiation/drug effects , Cell Line , Cell Self Renewal/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/transplantation , Phenotype , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Time Factors
15.
Oncotarget ; 7(32): 51193-51210, 2016 Aug 09.
Article in English | MEDLINE | ID: mdl-27323401

ABSTRACT

Hexavalent chromium [Cr(VI)] is a well-known human carcinogen associated with an increased risk of lung cancer. However, the mechanisms underlying Cr(VI)-induced carcinogenesis remain unclear. MicroRNA-21 (miR-21) is a key regulator of oncogenic processes. Studies have shown that miR-21 exerts its oncogenic activity by targeting the tumor suppressor gene programmed cell death 4 (PDCD4). The present study examined the role of miR-21-PDCD4 signaling in Cr(VI)-induced cell transformation and tumorigenesis. Results showed that Cr(VI) induces ROS generation in human bronchial epithelial (BEAS-2B) cells. Chronic exposure to Cr(VI) is able to cause malignant transformation in BEAS-2B cells. Cr(VI) caused a significant increase of miR-21 expression associated with an inhibition of PDCD4 expression. Notably, STAT3 transcriptional activation by IL-6 is crucial for the Cr(VI)-induced miR-21 elevation. Stable knockdown of miR-21 or overexpression of PDCD4 in BEAS-2B cells significantly reduced the Cr(VI)-induced cell transformation. Furthermore, the Cr(VI) induced inhibition of PDCD4 suppressed downstream E-cadherin protein expression, but promoted ß-catenin/TCF-dependent transcription of uPAR and c-Myc. We also found an increased miR-21 level and decreased PDCD4 expression in xenograft tumors generated with chronic Cr(VI)-exposed BEAS-2B cells. In addition, stable knockdown of miR-21 and overexpression of PDCD4 reduced the tumorogenicity of chronic Cr(VI)-exposed BEAS-2B cells in nude mice. Taken together, these results demonstrate that the miR-21-PDCD4 signaling axis plays an important role in Cr(VI)-induced carcinogenesis.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Bronchi/drug effects , Cell Transformation, Neoplastic/chemically induced , Chromium/toxicity , MicroRNAs/genetics , RNA-Binding Proteins/genetics , Reactive Oxygen Species/pharmacology , Respiratory Mucosa/drug effects , A549 Cells , Animals , Apoptosis Regulatory Proteins/metabolism , Bronchi/metabolism , Bronchi/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Humans , Lung Neoplasms/chemically induced , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/metabolism , RNA-Binding Proteins/metabolism , Reactive Oxygen Species/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Signal Transduction/drug effects , Signal Transduction/genetics
17.
J Environ Pathol Toxicol Oncol ; 34(1): 63-84, 2015.
Article in English | MEDLINE | ID: mdl-25746832

ABSTRACT

DNA methylation and histone modification promote opening and closure of chromatin structure, which affects gene expression without altering the DNA sequence. Epigenetic markers regulate the dynamic nature of chromatin structure at different levels: DNA, histone, noncoding RNAs, as well as the higher-order chromatin structure. Accumulating evidence strongly suggests that arsenic-induced carcinogenesis involves frequent changes in the epigenetic marker. However, progress in identifying arsenic-induced epigenetic changes has already been made using genome-wide approaches; the biological significance of these epigenetic changes remains unknown. Moreover, arsenic-induced changes in the chromatin state alter gene expression through the epigenetic mechanism. The current review provides a summary of recent literature regarding epigenetic changes caused by arsenic in carcinogenesis. We highlight the transgenerational studies needed to explicate the biological significance and toxicity of arsenic over a broad spectrum.


Subject(s)
Arsenic/toxicity , Epigenesis, Genetic , Animals , Carcinogenesis , DNA Methylation , Histones/metabolism , Humans , Mice , MicroRNAs/physiology , Promoter Regions, Genetic
19.
Toxicol Sci ; 130(2): 269-80, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22872060

ABSTRACT

Health effects due to environmental exposure to arsenic are a major global health concern. Arsenic has been known to induce carcinogenesis and enhance tumor development via complex and unclear mechanism. Ethanol is also a well-established risk factor for many malignancies. However, little is known about the effects of coexposure to arsenic and ethanol in tumor development. In this study, we investigate the signaling and angiogenic effect of coexposure of arsenic and ethanol on different colon cancer cell lines. Results show that ethanol markedly enhanced arsenic-induced tumor angiogenesis in vitro. These responses are related to intracellular reactive oxygen species (ROS) generation, NADPH oxidase activation, and upregulation of PI3K/Akt and hypoxia-inducible factor 1 alpha (HIF-1α) signaling. We have also found that ethanol increases the arsenic-induced expression and secretion of angiogenic signaling molecules such as vascular endothelial growth factor, which further confirmed the above observation. Antioxidant enzymes inhibited arsenic/ethanol-induced tumor angiogenesis, demonstrating that the responsive signaling pathways of coexposure to arsenic and ethanol are related to ROS generation. We conclude that ethanol is able to enhance arsenic-induced tumor angiogenesis in colorectal cancer cells via the HIF-1α pathway. These results indicate that alcohol consumption should be taken into consideration in the investigation of arsenic-induced carcinogenesis in arsenic-exposed populations.


Subject(s)
Arsenites/toxicity , Carcinogens/toxicity , Colonic Neoplasms/metabolism , Ethanol/toxicity , Human Umbilical Vein Endothelial Cells/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neovascularization, Physiologic/drug effects , Paracrine Communication , Sodium Compounds/toxicity , Catalase/metabolism , Cell Survival/drug effects , Colonic Neoplasms/blood supply , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Culture Media, Conditioned/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Genes, Reporter , HCT116 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , NADPH Oxidases/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Superoxide Dismutase/metabolism , Time Factors , Transfection , Up-Regulation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
20.
Toxicol Sci ; 119(2): 335-45, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21047991

ABSTRACT

Occupational exposure to chromium (Cr) compounds has been shown to cause serious toxic and carcinogenic effects. The skin is an important target for the compounds in industrially exposed Cr workers. c-Jun NH(2)-terminal kinase (JNK) regulates cell proliferation, apoptosis, and differentiation. This protein's effects on cellular response depend upon the cell type and stimuli. The mechanisms by which hexavalent chromium (Cr(VI)) leads to apoptosis in the skin are unclear at present. The aim of this study is to examine whether JNK regulates apoptosis in Cr(VI)-exposed mouse JB6 epidermal cells. The present study showed that Cr(VI) induced apoptotic cell death through JNK activation. The blockage of JNK by small interference RNA (si-RNA) transfection suppressed Cr(VI)-induced apoptotic cell death with the concomitant downregulation of antiapoptotic Bcl-2 family proteins, mitochondrial membrane depolarization (Δψm), caspase activation, and poly (ADP-ribose) polymerase cleavage. However, inhibition of c-Jun expression by si-RNA transfection enhanced cytotoxicity, which corresponded to increasing apoptosis and Δψm. This phenomenon is associated with p53 activation caused by increasing reactive oxygen species (ROS) levels because of the downregulation of superoxide dismutase expression in si-c-Jun-transfected cells. Taken together, Cr(VI) induces apoptosis via JNK-mediated signaling, whereas c-Jun activation acts as an inhibitor of apoptotic signaling. Additionally, ROS generated by Cr(VI) is a pivotal regulator of JNK.


Subject(s)
Apoptosis/drug effects , Chromium/toxicity , JNK Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , Animals , Blotting, Western , Cell Line , Electron Spin Resonance Spectroscopy , Mice , Reactive Oxygen Species/metabolism , Superoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL