Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 506
Filter
1.
Nat Commun ; 13(1): 136, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013263

ABSTRACT

Emerging research supports that triclosan (TCS), an antimicrobial agent found in thousands of consumer products, exacerbates colitis and colitis-associated colorectal tumorigenesis in animal models. While the intestinal toxicities of TCS require the presence of gut microbiota, the molecular mechanisms involved have not been defined. Here we show that intestinal commensal microbes mediate metabolic activation of TCS in the colon and drive its gut toxicology. Using a range of in vitro, ex vivo, and in vivo approaches, we identify specific microbial ß-glucuronidase (GUS) enzymes involved and pinpoint molecular motifs required to metabolically activate TCS in the gut. Finally, we show that targeted inhibition of bacterial GUS enzymes abolishes the colitis-promoting effects of TCS, supporting an essential role of specific microbial proteins in TCS toxicity. Together, our results define a mechanism by which intestinal microbes contribute to the metabolic activation and gut toxicity of TCS, and highlight the importance of considering the contributions of the gut microbiota in evaluating the toxic potential of environmental chemicals.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Carcinogens/antagonists & inhibitors , Colitis/prevention & control , Colorectal Neoplasms/prevention & control , Glucuronidase/antagonists & inhibitors , Glycoside Hydrolase Inhibitors/pharmacology , Triclosan/antagonists & inhibitors , Animals , Anti-Infective Agents, Local/chemistry , Anti-Infective Agents, Local/metabolism , Anti-Infective Agents, Local/toxicity , Anticarcinogenic Agents/chemistry , Anticarcinogenic Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Biotransformation , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogens/chemistry , Carcinogens/metabolism , Carcinogens/toxicity , Colitis/chemically induced , Colitis/enzymology , Colitis/microbiology , Colon/drug effects , Colon/microbiology , Colon/pathology , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/microbiology , Gastrointestinal Microbiome/drug effects , Gene Expression , Glucuronidase/chemistry , Glucuronidase/genetics , Glucuronidase/metabolism , Glycoside Hydrolase Inhibitors/chemistry , Humans , Mice , Mice, Inbred C57BL , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Triclosan/chemistry , Triclosan/metabolism , Triclosan/toxicity
2.
Comput Math Methods Med ; 2021: 4883509, 2021.
Article in English | MEDLINE | ID: mdl-34956398

ABSTRACT

Accumulating studies revealed association between development of glioma and miRNA dysregulation. A case in point is miR-381-3p, but its mechanism in glioma is unclear yet. In this work, we confirmed that overexpressed miR-381-3p repressed biological functions of glioma cells. Additionally, we also discovered that upregulated anthrax toxin receptor 1 (ANTXR1) was negatively mediated by miR-381-3p. We further proved that miR-381-3p-targeted ANTXR1 was able to counteract the suppression of miR-381-3p on biological functions of glioma. We concluded that miR-381-3p and ANTXR1 were both important factors in modulating glioma progression. miR-381-3p/ANTXR1 axis is expected to be a molecular target for glioma.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , MicroRNAs/genetics , Microfilament Proteins/antagonists & inhibitors , Receptors, Cell Surface/antagonists & inhibitors , Carcinogens/antagonists & inhibitors , Cell Line, Tumor , Computational Biology , Disease Progression , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Microfilament Proteins/genetics , Receptors, Cell Surface/genetics , Up-Regulation
3.
Food Chem Toxicol ; 157: 112582, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34582963

ABSTRACT

The present study investigated the protective effects and mechanism of action of cyanidin-3-O-glucoside (C3G) and its major metabolite protocatechuic acid (PCA) against 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) induced cytotoxicity in HepG2 cells. The results demonstrated that C3G and PCA dose-dependently suppressed PhIP-induced mutation in Salmonella typhimurium TA98, and inhibited PhIP-induced cytotoxicity and apoptosis in HepG2 cells. Western blot analysis indicated that C3G and PCA minimized PhIP-induced cell damage by reversing the abnormal expression of Bax/Bcl-2, Cytochrome c, cleaved Caspase-3, XIAP, Nrf2, HO-1, LC3 and p62 involved in intrinsic apoptotic and Nrf2/p62 pathways. Molecular docking results revealed that C3G and PCA were able to interfere with Nrf2 signaling and apoptotic cascade through binding to Keap1 and Bcl-2. Moreover, the protective effect of C3G was stronger than that of PCA. These findings suggested that dietary consumption of food sources rich in C3G can fight against the health risks of heterocyclic aromatic amines.


Subject(s)
Anthocyanins/pharmacology , Apoptosis/drug effects , Carcinogens/toxicity , Glycosides/pharmacology , Hep G2 Cells/drug effects , Hydroxybenzoates/pharmacology , Imidazoles/toxicity , NF-E2-Related Factor 2/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction/drug effects , Carcinogens/antagonists & inhibitors , Hep G2 Cells/metabolism , Humans , Imidazoles/antagonists & inhibitors , Molecular Docking Simulation
4.
Anticancer Agents Med Chem ; 21(16): 2100-2110, 2021 10 28.
Article in English | MEDLINE | ID: mdl-33573577

ABSTRACT

BACKGROUND: Sanguinarine, a kind of benzophenanthridine alkaloid, is a natural compound with potential development value for its anticancer activity. Hundreds of studies have been carried out in vivo or in vitro, trying to make it feasible for the anticancer clinic medication of sanguinarine. However, sanguinarine was branded as a toxicant or even a carcinogen according to some toxicological experiments and cancer investigations. OBJECTIVE: Aiming at safety and effectiveness of sanguinarine, this article reviews the extant information on present studies of sanguinarine, and both anticancer carcinogenesis mechanism details are summarized. The future potential research directions of this valued compound are also discussed to provide reference for future drugs development. METHODS: PubMed, Web of Science, CNKI and WanFang databases were used to search current studies and experimental researches about anticancer effect or carcinogenic information of sanguinarine. RESULTS: Our results indicated that sanguinarine exhibited anticancer effect through anti-proliferation, anti-invasion, anti-angiogenesis and apoptosis within cancer lesion. Also, many clinical investigations indicated that sanguinarine and its related products might be associated to pre-carcinoma within oral or skin potentially. CONCLUSION: Sanguinarine is a natural compound with good development value for its potent anticancer activity; however, its carcinogenesis effect should also be taken seriously. Studies on structural modification and analogue design should be carried out to improve its safety and efficiency in the future.


Subject(s)
Antineoplastic Agents/pharmacology , Benzophenanthridines/pharmacology , Carcinogens/antagonists & inhibitors , Isoquinolines/pharmacology , Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Benzophenanthridines/chemistry , Cell Proliferation/drug effects , Humans , Isoquinolines/chemistry , Neoplasms/pathology
5.
Int J Biol Macromol ; 176: 145-156, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33571591

ABSTRACT

Karyopherins mediate the macromolecular transport between the cytoplasm and the nucleus and participate in cancer progression. However, the role and mechanism of importin-11 (IPO11), a member of the karyopherin family, in glioma progression remain undefined. Effects of IPO11 on glioma progression were detected using CCK-8, colony formation assay, flow cytometry analysis, caspase-3 activity assay, and Transwell invasion assay. Western blot analysis was used to detect the expression of active caspase-3, active caspase-7, active caspase-9, N-cadherin, Vimentin, E-cadherin, ß-catenin, and c-Myc. The activity of Wnt/ß-catenin pathway was evaluated by the T-cell factor/lymphoid enhancer factor (TCF/LEF) transcription factor reporter assay. Results showed that IPO11 knockdown inhibited proliferation and reduced colony number in glioma cells. IPO11 silencing promoted the apoptotic rate, increased expression levels of active caspase-3, caspase-7, and caspase-9, and enhanced caspase-3 activity. Moreover, IPO11 silencing inhibited glioma cell invasion by suppressing epithelial-to-mesenchymal transition (EMT). Mechanistically, IPO11 knockdown inactivated the Wnt/ß-catenin pathway. ß-Catenin overexpression abolished the effects of IPO11 silencing on the proliferation, apoptosis, and invasion in glioma cells. Furthermore, IPO11 silencing blocked the malignant phenotypes and repressed the Wnt/ß-catenin pathway in vivo. In conclusion, IPO11 knockdown suppressed the malignant phenotypes of glioma cells by inactivating the Wnt/ß-catenin pathway.


Subject(s)
Brain Neoplasms/metabolism , Carcinogens/metabolism , Glioma/metabolism , beta Karyopherins/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Carcinogens/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Female , Gene Knockdown Techniques , Glioma/genetics , Glioma/pathology , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , Phenotype , Transcriptome , Up-Regulation , Wnt Signaling Pathway , beta Catenin/genetics , beta Catenin/metabolism , beta Karyopherins/antagonists & inhibitors , beta Karyopherins/genetics
6.
Curr Cancer Drug Targets ; 21(1): 21-54, 2021.
Article in English | MEDLINE | ID: mdl-33023449

ABSTRACT

CYP1A1 and CYP1B1 are extrahepatic P450 family members involved in the metabolism of procarcinogens, such as PAHs, heterocyclic amines and halogen-containing organic compounds. CYP1A1/1B1 also participate in the metabolism of endogenous 17-ß-estradiol, producing estradiol hydroquinones, which are the intermediates of carcinogenic semiquinones and quinones. CYP1A1 and CYP1B1 proteins share approximately half amino acid sequence identity but differ in crystal structures. As a result, CYP1A1 and CYP1B1 have different substrate specificity to chemical procarcinogens. This review will introduce the general molecular biology knowledge of CYP1A1/1B1 and the metabolic processes of procarcinogens regulated by these two enzymes. Over the last four decades, a variety of natural products and synthetic compounds which interact with CYP1A1/1B1 have been identified as effective chemo-preventive agents against chemical carcinogenesis. These compounds are mainly classified as indirect or direct CYP1A1/1B1 inhibitors based on their distinct mechanisms. Indirect CYP1A1/1B1 inhibitors generally impede the transcription and translation of CYP1A1/1B1 genes or interfere with the translocation of aryl hydrocarbon receptor (AHR) from the cytosolic domain to the nucleus. On the other hand, direct inhibitors inhibit the catalytic activities of CYP1A1/1B1. Based on the structural features, the indirect inhibitors can be categorized into the following groups: flavonoids, alkaloids and synthetic aromatics, whereas the direct inhibitors can be categorized into flavonoids, coumarins, stilbenes, sulfur containing isothiocyanates and synthetic aromatics. This review will summarize the in vitro and in vivo activities of these chemo-preventive agents, their working mechanisms, and related SARs. This will provide a better understanding of the molecular mechanism of CYP1 mediated carcinogenesis and will also give great implications for the discovery of novel chemo-preventive agents in the near future.


Subject(s)
Carcinogenesis , Carcinogens , Cytochrome P-450 CYP1A1 , Cytochrome P-450 CYP1B1 , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Carcinogenesis/chemically induced , Carcinogenesis/drug effects , Carcinogens/antagonists & inhibitors , Carcinogens/chemistry , Carcinogens/metabolism , Chemoprevention/methods , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1B1/genetics , Cytochrome P-450 CYP1B1/metabolism , Humans , Organic Chemistry Phenomena , Structure-Activity Relationship
7.
Eur J Pharmacol ; 886: 173445, 2020 Nov 05.
Article in English | MEDLINE | ID: mdl-32758571

ABSTRACT

The incidence of colon cancer increased worldwide in 2019 and its treatment is urgent from a quality of life perspective. A relationship has been reported between elevated numbers of tumor-associated macrophages (TAMs) in the tumor microenvironment and a poor prognosis in cancer patients, and M2 TAMs have been shown to promote tumor growth by immunosuppression through the stimulation of programmed death-1 (PD-1, an immune check point receptor), interleukin (IL)-1ß, and monocyte chemoattractant protein (MCP)-1. We herein examined the effects of three synthetic dihydroxystilbenes (2,3-, 3,4-, and 4,4'-dihydroxystilbenes) on colon carcinogenesis, colon tumor growth, and colon cytokines (IL-1ß, IL-6, and tumor necrosis factor (TNF)-α), a chemokine (MCP-1), vascular endothelial growth factor (VEGF), and PD-1 levels in azoxymethane (AOM) plus dextran sulfate sodium (DSS)-treated C57BL/6J mice. The three dihydroxystilbenes inhibited colon carcinogenesis and tumor growth as well as increases in colon IL-1ß, IL-6, MCP-1, and PD-1 levels in AOM/DDS-treated mice (in vivo). The three dihydroxystilbenes also suppressed COX-2 expression in colon tumors (in vivo). The results obtained also revealed that the three dihydroxystilbenes inhibited PD-1 elevations in M2-THP-1 macrophages (in vitro). Therefore, the inhibition of AOM/DSS-induced colon carcinogenesis and colon tumor growth by 2,3-, 3,4-, and 4,4'-dihydroxystilbenes appears to be due to the suppression of M2 TAM differentiation and activation and PD-1 expression (immunosuppression) via reductions in COX-2 expression levels in the colon tumor microenvironment.


Subject(s)
Apoptosis/drug effects , Chemokines/antagonists & inhibitors , Colonic Neoplasms/chemically induced , Colonic Neoplasms/prevention & control , Cytokines/antagonists & inhibitors , Dihydrostilbenoids/therapeutic use , Animals , Azoxymethane , Carcinogens/antagonists & inhibitors , Chemokine CCL2/drug effects , Colonic Neoplasms/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Dextran Sulfate , Dihydrostilbenoids/chemical synthesis , Dihydrostilbenoids/chemistry , Humans , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Tumor Microenvironment/drug effects
8.
J Natl Cancer Inst ; 112(1): 30-37, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31498409

ABSTRACT

The Monographs produced by the International Agency for Research on Cancer (IARC) apply rigorous procedures for the scientific review and evaluation of carcinogenic hazards by independent experts. The Preamble to the IARC Monographs, which outlines these procedures, was updated in 2019, following recommendations of a 2018 expert advisory group. This article presents the key features of the updated Preamble, a major milestone that will enable IARC to take advantage of recent scientific and procedural advances made during the 12 years since the last Preamble amendments. The updated Preamble formalizes important developments already being pioneered in the Monographs program. These developments were taken forward in a clarified and strengthened process for identifying, reviewing, evaluating, and integrating evidence to identify causes of human cancer. The advancements adopted include the strengthening of systematic review methodologies; greater emphasis on mechanistic evidence, based on key characteristics of carcinogens; greater consideration of quality and informativeness in the critical evaluation of epidemiological studies, including their exposure assessment methods; improved harmonization of evaluation criteria for the different evidence streams; and a single-step process of integrating evidence on cancer in humans, cancer in experimental animals, and mechanisms for reaching overall evaluations. In all, the updated Preamble underpins a stronger and more transparent method for the identification of carcinogenic hazards, the essential first step in cancer prevention.


Subject(s)
Carcinogens/antagonists & inhibitors , Neoplasms/prevention & control , Animals , Humans , International Agencies/organization & administration , Motivation , Program Evaluation , Public Health Surveillance
9.
Prostate ; 79(14): 1611-1621, 2019 10.
Article in English | MEDLINE | ID: mdl-31348843

ABSTRACT

BACKGROUND: The transcription factor signal transducer and activator of transcription 3 (STAT3) is implicated in cancer drug resistance, metastasis, and immunosuppression and has been identified as a promising therapeutic target for new anticancer drugs. Myeloid-derived suppressor cells (MDSCs) play a major role in the suppression of antitumor immunity and STAT3 is involved in the accumulation, generation, and function of MDSCs. Thus, targeting STAT3 holds the potential of reversing immunosuppression in cancer. This study aims to investigate the effect of the small molecule STAT3 inhibitor galiellalactone on prostate cancer cell- induced generation of MDSCs from monocytes and the effect on immunosuppressive factors and inflammatory cytokines. METHODS: Primary human monocytes were cocultured with prostate cancer cells (DU145, PC3, and LNCaP-IL6) or with conditioned medium (CM) from prostate cancer cells in the presence or absence of the STAT3 inhibitor galiellalactone. Monocytes were analyzed by flow cytometry for an MDSC-like phenotype (CD14+ HLA-DR-/lo ). The secretion and gene expression of immunosuppressive factors and inflammatory cytokines from prostate cancer cells and monocytes were investigated. RESULTS: Galiellalactone blocked the prostate cancer cell-induced generation of MDSC-like monocytes with an immunosuppressive phenotype ex vivo. Monocytes cultured with CM from prostate cancer cells showed increased expression of phosphorylated STAT3. Prostate cancer cells increased the expression of interleukin1ß (IL1ß), IL10, and IL6 in monocytes which was inhibited by galiellalactone. In addition, galiellalactone decreased indoleamine 2,3-dioxygenase gene expression in monocytes. Galiellalactone reduced the levels of IL8 and granulocyte macrophage-colony stimulating factor in prostate cancer cells per se. CONCLUSION: The STAT3 inhibitor galiellalactone may prevent the prostate cancer cell-induced generation of MDSCs and reverse the immunosuppressive mechanisms caused by the interplay between prostate cancer cells and MDSCs. This is a potential new immunotherapeutic approach for the treatment of prostate cancer.


Subject(s)
Carcinogens/antagonists & inhibitors , Immunosuppressive Agents/antagonists & inhibitors , Lactones/pharmacology , Myeloid-Derived Suppressor Cells/drug effects , Prostatic Neoplasms/pathology , STAT3 Transcription Factor/antagonists & inhibitors , Carcinogenesis/drug effects , Carcinogens/metabolism , Coculture Techniques , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Humans , Immunosuppressive Agents/metabolism , Male , Monocytes/drug effects , Monocytes/metabolism , Monocytes/pathology , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , Prostatic Neoplasms/metabolism , Tumor Cells, Cultured
10.
FASEB J ; 33(3): 3198-3211, 2019 03.
Article in English | MEDLINE | ID: mdl-30379590

ABSTRACT

Growth factor receptor-binding protein 10 (GRB10) is a well-known adaptor protein and a recently identified substrate of the mammalian target of rapamycin (mTOR). Depletion of GRB10 increases insulin sensitivity and overexpression suppresses PI3K/Akt signaling. Because the major reason for the limited efficacy of PI3K/Akt-targeted therapies in prostate cancer (PCa) is loss of mTOR-regulated feedback suppression, it is therefore important to assess the functional importance and regulation of GRB10 under these conditions. On the basis of these background observations, we explored the status and functional impact of GRB10 in PCa and found maximum expression in phosphatase and tensin homolog (PTEN)-deficient PCa. In human PCa samples, GRB10 inversely correlated with PTEN and positively correlated with pAKT levels. Knockdown of GRB10 in nontumorigenic PTEN null mouse embryonic fibroblasts and tumorigenic PCa cell lines reduced Akt phosphorylation and selectively activated a panel of receptor tyrosine kinases. Similarly, overexpression of GRB10 in PTEN wild-type PCa cell lines accelerated tumorigenesis and induced Akt phosphorylation. In PTEN wild-type PCa, GRB10 overexpression promoted mediated PTEN interaction and degradation. PI3K (but not mTOR) inhibitors reduced GRB10 expression, suggesting primarily PI3K-driven regulation of GRB10. In summary, our results suggest that GRB10 acts as a major downstream effector of PI3K and has tumor-promoting effects in prostate cancer.-Khan, M. I., Al Johani, A., Hamid, A., Ateeq, B., Manzar, N., Adhami, V. M., Lall, R. K., Rath, S., Sechi, M., Siddiqui, I. A., Choudhry, H., Zamzami, M. A., Havighurst, T. C., Huang, W., Ntambi, J. M., Mukhtar, H. Proproliferatve function of adaptor protein GRB10 in prostate carcinoma.


Subject(s)
GRB10 Adaptor Protein/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Animals , Carcinogens/antagonists & inhibitors , Carcinogens/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Fibroblasts/cytology , Fibroblasts/metabolism , GRB10 Adaptor Protein/antagonists & inhibitors , GRB10 Adaptor Protein/genetics , Gene Knockdown Techniques , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Models, Biological , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/genetics , RNA, Messenger , Signal Transduction
11.
J Vet Med Sci ; 80(8): 1301-1304, 2018 Aug 22.
Article in English | MEDLINE | ID: mdl-29937461

ABSTRACT

Cigarette smoke is a strong and independent risk factor for esophageal cancer, while the consumption of cow's milk has been proposed as a protective factor. The mechanistic role of milk in preventing cancer, however, has not been clarified. We focused our study on acrolein, an abundant unsaturated aldehyde present in cigarette smoke. Acrolein is a highly toxic compound and a putative carcinogen. Using a cell culture system, we found that (1) acrolein caused necrosis in Ramos Burkitt's lymphoma cells, (2) the necrosis was inhibited by preincubation of acrolein with milk, and (3) acrolein formed adducts with milk proteins. These results indicated the protective effects of cow's milk against acrolein-induced cytotoxicity via protein-acrolein adduct formation.


Subject(s)
Acrolein/antagonists & inhibitors , Carcinogens/antagonists & inhibitors , Milk Proteins/chemistry , Smoking/adverse effects , Animals , Cattle , Female , Male , Milk/chemistry
12.
Environ Sci Pollut Res Int ; 25(10): 10080-10089, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29383641

ABSTRACT

Cd is a hazardous substance and carcinogen that is present in the environment; it is known to cause toxic effects in living organisms. Sulforaphane is a naturally available phytochemical with antioxidant, anti-inflammatory, and anticarcinogenic properties. However, the effects of sulforaphane on Cd toxicity in human mesenchymal stem cells (hMSCs) are unknown. In the present study, we investigated the molecular mechanisms of the effects of sulforaphane on Cd toxicity in hMSCs by using MTT assays, acridine orange/ethidium bromide staining, Hoechst staining, LysoRed staining, assessment of mitochondrial membrane potential, and gene expression analysis. Cd decreased hMSC viability in a dose-dependent manner with an IC50 value of 56.5 µM. However, sulforaphane did not induce any significant reduction in cell viability. Nuclear morphological analysis revealed that Cd induced necrotic cell death. Additionally, Cd caused mitochondrial membrane potential loss in hMSCs. The treatment of Cd-exposed cells with sulforaphane (Cd-sulforaphane co-treatment) resulted in a significant recovery of the cell viability and nuclear morphological changes compared with that of cells treated with Cd only. The gene expression pattern of cells co-treated with Cd-sulforaphane was markedly different from that of Cd-treated cells, owing to the reduction in Cd toxicity. Our results clearly indicated that sulforaphane reduced Cd-induced toxic effects in hMSCs. Overall, the results of our study suggested that sulforaphane-rich vegetables and fruits can help to improve human health through amelioration of the molecular effects of Cd poisoning.


Subject(s)
Cadmium/toxicity , Carcinogens/toxicity , Isothiocyanates/pharmacology , Mesenchymal Stem Cells/drug effects , Protective Agents/pharmacology , Antioxidants/pharmacology , Carcinogens/antagonists & inhibitors , Cell Survival/drug effects , Cells, Cultured , Gene Expression/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Sulfoxides
13.
Curr Pharm Des ; 24(5): 595-614, 2018.
Article in English | MEDLINE | ID: mdl-29278208

ABSTRACT

BACKGROUND: Potassium bromate (KBrO3), a food additive, has been used in many bakery products as an oxidizing agent. It has been shown to induce renal cancer in many in-vitro and in-vivo experimental models. OBJECTIVES: This study evaluated the carcinogenic potential of potassium bromate (KBrO3) and the chemopreventive mechanisms of the anti-oxidant and anti-inflammatory phytochemical, curcumin against KBrO3-induced carcinogenicity. METHOD: Lactate dehydrogenase (LDH) cytotoxicity assay and morphological characteristics were used to assess curcumin's cytoprotective potential against KBrO3 toxicity. To assess the chemopreventive potential of curcumin against KBrO3-induced oxidative insult, intracellular H2O2 and the nuclear concentration of the DNA adduct 8- OHdG were measured. PCR array, qRT-PCR, and western blot analysis were used to identify dysregulated genes by KBrO3 exposure. Furthermore, immunofluorescence was used to evaluate the ciliary loss and the disturbance of cellular tight junction induced by KBrO3. RESULTS: Oxidative stress assays showed that KBrO3 increased the levels of intracellular H2O2 and the DNA adduct 8-OHdG. Combination of curcumin with KBrO3 efficiently reduced the level of H2O2 and 8-OHdG while upregulating the expression of catalase. PCR array, qRT-PCR, and western blot analysis revealed that KBrO3 dysregulated multiple genes involved in inflammation, proliferation, and apoptosis, namely CTGF, IL-1, and TRAF3. Moreover, qRT-PCR and immunofluorescence studies showed that KBrO3 negatively affected the tight junctional protein (ZO-1) and induced a degeneration of primary ciliary proteins. The negative impact of KBrO3 on cilia was markedly repressed by curcumin. CONCLUSION: Curcumin could potentially be used as a protective agent against carcinogenicity of KBrO3.


Subject(s)
Bromates/antagonists & inhibitors , Carcinogens/antagonists & inhibitors , Curcumin/pharmacology , Food Additives/adverse effects , Protective Agents/pharmacology , Bromates/pharmacology , Carcinogens/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Curcumin/analysis , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Humans , Protective Agents/analysis
14.
BMC Pharmacol Toxicol ; 18(1): 17, 2017 04 22.
Article in English | MEDLINE | ID: mdl-28431577

ABSTRACT

BACKGROUND: The present study was designed to investigate the protective effect of aqueous date extract (ADE) against the dichloroacetic acid (DCA)-induced testicular injury in rats. METHODS: Forty-eight male Wistar rats were randomly divided into six groups of eight: group I served as the control; group II was given ADE (4 ml/kg) by gavage; groups III and IV received DCA at 0.5 and 2 g/L drinking water, respectively; and groups V and VI received DCA at 0.5 and 2 g/L drinking water, respectively, before ADE administration. The experiment was performed for two months. RESULTS: Results showed that the absolute weights of testes and epididymis were decreased following the DCA administration. The testosterone, FSH and LH levels were also decreased. Severe histopathological changes in testes were observed including degeneration of seminiferous tubules and depletion of germ cells. These changes were associated with alterations of oxidative stress markers. Levels of lipid peroxidation and SOD and CAT activities were increased, while activity of GPx and GSH levels were decreased. Pretreatment with ADE has effectively alleviated the oxidative stress induced by DCA thereby restoring these parameters to normal values. CONCLUSIONS: These results suggest that ADE has a protective effect over DCA-induced oxidative damage in rat testes.


Subject(s)
Dichloroacetic Acid/toxicity , Disinfectants/toxicity , Phoeniceae/chemistry , Plant Extracts/therapeutic use , Testis/drug effects , Animals , Antioxidants/metabolism , Carcinogens/antagonists & inhibitors , Carcinogens/toxicity , Dichloroacetic Acid/antagonists & inhibitors , Disinfectants/antagonists & inhibitors , Epididymis/drug effects , Gonadal Steroid Hormones/blood , Lipid Peroxidation/drug effects , Male , Organ Size/drug effects , Oxidative Stress/drug effects , Protective Agents/therapeutic use , Rats , Rats, Wistar , Testis/pathology
15.
J Appl Toxicol ; 37(4): 417-425, 2017 04.
Article in English | MEDLINE | ID: mdl-27581495

ABSTRACT

A body of epidemiological evidence implicates exposure to endocrine disrupting chemicals (EDCs) with increased susceptibility to breast cancer. To evaluate the physiological effects of a suspected EDC in vivo, we exposed MCF-7 breast cancer cells and a patient-derived xenograft (PDX, estrogen receptor positive) to physiological levels of methylparaben (mePB), which is commonly used in personal care products as a preservative. mePB pellets (4.4 µg per day) led to increased tumor size of MCF-7 xenografts and ER+ PDX tumors. mePB has been thought to be a xenoestrogen; however, in vitro exposure of 10 nM mePB failed to increase MCF-7 cell proliferation or induction of canonical estrogen-responsive genes (pS2 and progesterone receptor), in contrast to 17ß-estradiol (E2) treatment. MCF-7 and PDX-derived mammospheres exhibited increased size and up-regulation of canonical stem cell markers ALDH1, NANOG, OCT4 and SOX2 when exposed to mePB; these effects were not observed for MDA-MB-231 (ER- ) mammospheres. As tumor-initiating cells (TICs) are also believed to be responsible for chemoresistance, mammospheres were treated with either tamoxifen or the pure anti-estrogen fulvestrant in the presence of mePB. Blocking the estrogenic response was not sufficient to block NANOG expression in mammospheres, pointing to a non-classic estrogen response or an ER-independent mechanism of mePB promotion of mammosphere activity. Overall, these results suggest that mePB increases breast cancer tumor proliferation through enhanced TIC activity, in part via regulation of NANOG, and that mePB may play a direct role in chemoresistance by modulating stem cell activity. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Breast Neoplasms/chemically induced , Breast Neoplasms/genetics , Carcinogens/toxicity , Endocrine Disruptors/toxicity , Neoplastic Stem Cells/drug effects , Parabens/toxicity , Receptors, Estrogen/genetics , Animals , Carcinogens/antagonists & inhibitors , Cell Proliferation/drug effects , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Female , Humans , MCF-7 Cells , Mice , Mice, Nude , Neoplasm Proteins/genetics , Ovariectomy , Xenograft Model Antitumor Assays
16.
Biochem Pharmacol ; 117: 35-45, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27520485

ABSTRACT

The neoplastic transformation of cells and inflammation are processes that contribute to tumor initiation. Recently, emerging evidence has suggested that epigenetic alterations are also implicated in the early stages of carcinogenesis. Therefore, potent small molecules targeting epigenetic regulators have been developed as novel cancer therapeutic and preventive strategies. Bromodomain and extraterminal domain (BET) proteins are epigenetic readers that play key roles at the interface between chromatin modification and transcriptional regulation. In this study, we investigated the effect of the BET inhibitor JQ-1 on malignant transformation induced by 12-O-tetradecanoylphorbol-13-acetate (TPA) in mouse skin epidermal JB6 P+ cells. Treatment with JQ-1 effectively impaired TPA-induced colony formation in vitro. At the molecular level, the expression of several key TPA-induced pro-survival and pro-proliferative genes (Bcl2, Cyclin D1, and c-Myc) decreased rapidly after BET inhibition. In addition, JQ-1 treatment attenuated the activation of inflammatory NF-κB signaling triggered by TPA. Luciferase reporter assays using plasmids carrying different elements from the COX2 or IL6 promoters demonstrated that JQ-1 does not directly inhibit interactions between NF-κB and its binding sequence; rather, it affects CRE-element-associated transcriptional enhancement. Through siRNA gene silencing, we found that JQ-1 inhibits the p300-dependent transcriptional activation of COX2, which correlates with the results of the luciferase assay. Chromatin immunoprecipitation assays showed that TPA elevated H3K27Ac enrichment in the COX2 promoter region, which is mediated by p300, and Brd4. JQ-1 treatment did not change H3K27Ac levels but decreased the recruitment of Brd4 and RNA Polymerase II. Collectively, our study reveals that the BET inhibitor JQ-1 exerts potent anti-cancer and anti-inflammatory effects by interfering with the core transcriptional program of neoplastic transformation.


Subject(s)
Anticarcinogenic Agents/pharmacology , Azepines/pharmacology , Cell Transformation, Neoplastic/drug effects , Epidermis/drug effects , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Nuclear Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Triazoles/pharmacology , Acetylation/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carcinogens/antagonists & inhibitors , Carcinogens/toxicity , Cell Line , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cyclooxygenase 2/chemistry , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , E1A-Associated p300 Protein/antagonists & inhibitors , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/metabolism , Epidermis/immunology , Epidermis/metabolism , Epidermis/pathology , Genes, Reporter/drug effects , Histones/metabolism , Humans , Mice , Nuclear Proteins/metabolism , Promoter Regions, Genetic/drug effects , Protein Processing, Post-Translational/drug effects , RNA Interference , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/antagonists & inhibitors , Tetradecanoylphorbol Acetate/toxicity , Transcription Factors/metabolism
17.
J Nutr Biochem ; 33: 54-62, 2016 07.
Article in English | MEDLINE | ID: mdl-27260468

ABSTRACT

Ursolic acid (UA), a well-known natural triterpenoid found in abundance in blueberries, cranberries and apple peels, has been reported to possess many beneficial health effects. These effects include anticancer activity in various cancers, such as skin cancer. Skin cancer is the most common cancer in the world. Nuclear factor E2-related factor 2 (Nrf2) is a master regulator of antioxidative stress response with anticarcinogenic activity against UV- and chemical-induced tumor formation in the skin. Recent studies show that epigenetic modifications of Nrf2 play an important role in cancer prevention. However, the epigenetic impact of UA on Nrf2 signaling remains poorly understood in skin cancer. In this study, we investigated the epigenetic effects of UA on mouse epidermal JB6 P+ cells. UA inhibited cellular transformation by 12-O-tetradecanoylphorbol-13-acetate at a concentration at which the cytotoxicity was no more than 25%. Under this condition, UA induced the expression of the Nrf2-mediated detoxifying/antioxidant enzymes heme oxygenase-1, NAD(P)H:quinone oxidoreductase 1 and UDP-glucuronosyltransferase 1A1. DNA methylation analysis revealed that UA demethylated the first 15 CpG sites of the Nrf2 promoter region, which correlated with the reexpression of Nrf2. Furthermore, UA reduced the expression of epigenetic modifying enzymes, including the DNA methyltransferases DNMT1 and DNMT3a and the histone deacetylases (HDACs) HDAC1, HDAC2, HDAC3 and HDAC8 (Class I) and HDAC6 and HDAC7 (Class II), and HDAC activity. Taken together, these results suggest that the epigenetic effects of the triterpenoid UA could potentially contribute to its beneficial effects, including the prevention of skin cancer.


Subject(s)
Anticarcinogenic Agents/metabolism , Cell Transformation, Neoplastic , Epidermis/metabolism , Epigenesis, Genetic , NF-E2-Related Factor 2/agonists , Skin Neoplasms/prevention & control , Triterpenes/metabolism , Animals , Anticarcinogenic Agents/adverse effects , Antioxidants/adverse effects , Antioxidants/metabolism , Carcinogens/antagonists & inhibitors , Carcinogens/toxicity , Cell Line , Cell Survival/drug effects , Cell Transformation, Neoplastic/drug effects , DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/drug effects , Dietary Supplements/adverse effects , Enzyme Repression/drug effects , Epidermal Cells , Epidermis/drug effects , Epigenesis, Genetic/drug effects , Histone Deacetylases/chemistry , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Mice , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Promoter Regions, Genetic/drug effects , Skin Neoplasms/chemically induced , Skin Neoplasms/metabolism , Triterpenes/adverse effects , Ursolic Acid
18.
Toxicol Lett ; 258: 93-100, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27329536

ABSTRACT

Endocannabinoids are synthetized as a results of demand from membrane phospholipids. The formation and actions of these lipid mediators depend to a great extent on the prevalence of precursor fatty acid (FA), and can be influenced by diet or supplementation. The purpose of this study was to evaluate the interactive effects of lipopolysaccharide (LPS) and benzo(a)pyrene (BaP) in RAW 264.7 cells supplemented with docosahexaenoic acid (DHA). After LPS and/or BaP treatment in macrophages pre-incubated with DHA, a significant decrease in the amount of fatty acid was observed. The highest content of monounsaturated fatty acids was detected in RAW 264.7 cells co-treated with LPS and BaP. Significant interactions between LPS and BaP co-treatment in terms of endocannabinoid levels were observed in RAW 264.7 cells after DHA supplementation. The highest amount of endocannabinoids was detected in macrophages supplemented with DHA and co-treated with BaP and LPS: arachidonoyl ethanolamine AEA (5.9µg/mL), docosahexaenoyl ethanolamide DHEA (10.6µg/mL) and nervonoyl ethanolamide NEA (16.5µg/mL). The highest expression of cyclooxygenase (COX-2) and cannabinoid receptor 2 (CB2) was noted in macrophages supplemented with DHA and activated with LPS and BaP. Our data suggested a novel, CB2 receptor-dependent, environmental stress reaction in macrophages co-treated with LPS and BaP after supplementation with DHA. Despite the synergistic LPS and BaP action DHA potentiates the anti-inflammatory response in RAW 264.7 cells.


Subject(s)
Benzo(a)pyrene/antagonists & inhibitors , Carcinogens/antagonists & inhibitors , Docosahexaenoic Acids/metabolism , Endocannabinoids/antagonists & inhibitors , Lipopolysaccharides/antagonists & inhibitors , Macrophage Activation/drug effects , Macrophages/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anticarcinogenic Agents/metabolism , Arachidonic Acids , Benzo(a)pyrene/toxicity , Carcinogens/toxicity , Cyclooxygenase 2/chemistry , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dietary Supplements , Drug Synergism , Endocannabinoids/metabolism , Fatty Acids, Monounsaturated/metabolism , Gene Expression Regulation/drug effects , Lipopolysaccharides/toxicity , Macrophages/immunology , Macrophages/metabolism , Mice , Polyunsaturated Alkamides , RAW 264.7 Cells , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/metabolism
19.
Lung Cancer ; 96: 7-14, 2016 06.
Article in English | MEDLINE | ID: mdl-27133742

ABSTRACT

RATIONALE: Nintedanib is a potent, triple angiokinase inhibitor of vascular endothelial growth factor, fibroblast growth factor, and platelet-derived growth factor, and has been recently approved for the treatment of non-small cell lung cancer (NSCLC), following first-line chemotherapy. It is well established that microenvironment plays an important role in tumor progression. Therefore, targeting tumor microenvironment-cancer cell interaction may provide a significant therapeutic target. In this study we tested the effect of Nintedanib on NSCLC cells directly and in the presence of normal and tumor soluble microenvironment. METHODS: Primary fibroblast cultures derived from NSCLC tumors and normal lung tissues were established and their supernatants were collected. These supernatants were added to NSCLC cell lines (H1299, H460 and A549) cultured with/without Nintedanib (0.1-10µM) for 24 and 48h. Cell death (AnnexinV-PI, flow-cytometry), cell number, proliferation (PCNA), protein expression (immunoblotting) and cell migration (scratch test), were tested. Expression of 10 pro-angiogenic cytokines was measured by ELISA-based quantitative array. RESULTS: Tumor and normal supernatants demonstrated similar pro-metastatic effects on the NSCLC phenotype: both elevated cancer cell number, PCNA levels, reduced total and apoptotic cell death and facilitated cell migration. Nintedanib had limited but significant effects on the NSCLC cell number, cell death and migration, but required high doses. However, at lower doses Nintedanib caused cell detachment and elevated integrin-alpha 5 and EGFR levels, both markers of anoikis resistance. This suggests them as possible targets in combination with Nintedanib. Moreover, Nintedanib completely blocked the supernatants ability to facilitate the aggressive cancer cell characteristics. While cytokine array analysis showed no significant changes in FGF, PDGF or VEGF, we found that both supernatants contained high HGF levels, suggesting it as the facilitator of cell migration and proliferation. CONCLUSION: Our results demonstrate that tumor microenvironment-cancer cell interaction is a therapeutic target and should be considered when new drugs are tested.


Subject(s)
Carcinogens/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Lung Neoplasms/drug therapy , Tumor Microenvironment/drug effects , A549 Cells , Angiogenesis Inhibitors/therapeutic use , Carcinogens/pharmacology , Carcinoma, Non-Small-Cell Lung/blood supply , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Lung/drug effects , Lung/pathology , Lung Neoplasms/blood supply , Lung Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Platelet-Derived Growth Factor , Vascular Endothelial Growth Factor A/antagonists & inhibitors
20.
Article in English | MEDLINE | ID: mdl-27052947

ABSTRACT

This work reports the application of banana peel as a novel bioadsorbent for in vitro removal of five mycotoxins (aflatoxins (AFB1, AFB2, AFG1, AFG2) and ochratoxin A). The effect of operational parameters including initial pH, adsorbent dose, contact time and temperature were studied in batch adsorption experiments. Scanning electron microscopy (SEM), Fourier transform infrared spectroscopy (FTIR) and point of zero charge (pHpzc) analysis were used to characterise the adsorbent material. Aflatoxins' adsorption equilibrium was achieved in 15 min, with highest adsorption at alkaline pH (6-8), while ochratoxin has not shown any significant adsorption due to surface charge repulsion. The experimental equilibrium data were tested by Langmuir, Freundlich and Hill isotherms. The Langmuir isotherm was found to be the best fitted model for aflatoxins, and the maximum monolayer coverage (Q0) was determined to be 8.4, 9.5, 0.4 and 1.1 ng mg(-1) for AFB1, AFB2, AFG1 and AFG2 respectively. Thermodynamic parameters including changes in free energy (ΔG), enthalpy (ΔH) and entropy (ΔS) were determined for the four aflatoxins. Free energy change and enthalpy change demonstrated that the adsorption process was exothermic and spontaneous. Adsorption and desorption study at different pH further demonstrated that the sorption of toxins was strong enough to sustain pH changes that would be experienced in the gastrointestinal tract. This study suggests that biosorption of aflatoxins by dried banana peel may be an effective low-cost decontamination method for incorporation in animal feed diets.


Subject(s)
Absorption, Physicochemical , Aflatoxins/antagonists & inhibitors , Carcinogens/antagonists & inhibitors , Dietary Fiber/analysis , Fruit/chemistry , Industrial Waste/analysis , Musa/chemistry , Adsorption , Aflatoxin B1/analysis , Aflatoxin B1/antagonists & inhibitors , Aflatoxin B1/chemistry , Aflatoxins/analysis , Aflatoxins/chemistry , Animal Feed/adverse effects , Animal Feed/analysis , Animal Feed/economics , Animal Feed/microbiology , Animals , Carcinogens/analysis , Carcinogens/chemistry , Dietary Fiber/economics , Dietary Fiber/therapeutic use , Food Contamination , Food-Processing Industry/economics , Foodborne Diseases/etiology , Foodborne Diseases/prevention & control , Foodborne Diseases/veterinary , Hydrogen-Ion Concentration , Industrial Waste/economics , Queensland , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...