Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Chem Res Toxicol ; 33(2): 625-633, 2020 02 17.
Article in English | MEDLINE | ID: mdl-31841318

ABSTRACT

DNA methylating agents are abundant in the environment and are sometimes used in cancer chemotherapy. They react with DNA to form methyl-DNA adducts and byproduct lesions that can be both toxic and mutagenic. Foremost among the mutagenic lesions is O6-methylguanine (m6G), which base pairs with thymine during replication to cause GC → AT mutations. The gpt delta C57BL/6J mouse strain of Nohmi et al. (Mol. Mutagen 1996, 28, 465-70) reliably produces mutational spectra of many DNA damaging agents. In this work, mouse embryo fibroblasts (MEFs) were made from gpt delta C57BL/6J mice and evaluated as a screening tool to determine the qualitative and quantitative features of mutagenesis by N-methyl-N-nitrosourea (MNU), a direct-acting DNA alkylator that serves as a model for environmental N-nitrosamines, such as N-nitrosodimethylamine and therapeutic agents such as Temozolomide. The DNA repair protein MGMT (O6-methylguanine DNA methyltransferase) protects against environmental mutagenesis by DNA methylating agents and, by removing m6G, limits the therapeutic potential of Temozolomide in cancer therapy. The gpt delta MEFs were treated with MNU to establish dose-dependent toxicity. In parallel, MNU mutagenicity was determined in the presence and absence of the MGMT inhibitor AA-CW236 (4-(2-(5-(chloromethyl)-4-(4-(trifluoromethoxy)phenyl)-1H-1,2,3-triazol-1-yl)ethyl)-3,5-dimethylisoxazole). With and without the inhibitor, the principal mutagenic event of MNU was GC → AT, but more mutations were observed when the inhibitor was present. Evidence that the mutagenic lesion was m6G was based on mass spectral data collected using O6-methyl-d3-guanine as an internal standard; m6G levels were higher in AA-CW236 treated MEFs by an amount proportional to the higher mutation frequency seen in the same cells. This work establishes gpt delta MEFs as a versatile tool for probing mutagenesis by environmental and therapeutic agents and as a cell culture model in which chemical genetics can be used to determine the impact of DNA repair on biological responses to DNA damaging agents.


Subject(s)
Alkylating Agents/pharmacology , DNA Modification Methylases/antagonists & inhibitors , DNA Repair Enzymes/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Methylnitrosourea/pharmacology , Mutagenesis/drug effects , Tumor Suppressor Proteins/antagonists & inhibitors , Alkylating Agents/chemistry , Animals , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Enzyme Inhibitors/chemistry , Fibroblasts/metabolism , Methylnitrosourea/chemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tumor Suppressor Proteins/metabolism
2.
Planta Med ; 83(9): 778-789, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28249303

ABSTRACT

Andrographis paniculata has been widely used in Scandinavian and Asian counties for the treatment of the common cold, fever, and noninfectious diarrhea. The present study was carried out to investigate the physiological effects of short-term multiple dose administration of a standardized A. paniculata capsule used for treatment of the common cold and uncomplicated upper respiratory tract infections, including blood pressure, electrocardiogram, blood chemistry, hematological profiles, urinalysis, and blood coagulation in healthy Thai subjects. Twenty healthy subjects (10 males and 10 females) received 12 capsules per day orally of 4.2 g of a standardized A. paniculata crude powder (4 capsules of 1.4 g of A. paniculata, 3 times per day, 8 h intervals) for 3 consecutive days. The results showed that all of the measured clinical parameters were found to be within normal ranges for a healthy person. However, modulation of some parameters was observed after the third day of treatment, for example, inductions of white blood cells and absolute neutrophil count in the blood, a reduction of plasma alkaline phosphatase, and an induction of urine pH. A rapid and transient reduction in blood pressure was observed at 30 min after capsule administration, resulting in a significant reduction of mean systolic blood pressure. There were no serious adverse events observed in the subjects during the treatment period. In conclusion, this study suggests that multiple oral dosing of A. paniculata at the normal therapeutic dose for the common cold and uncomplicated upper respiratory tract infections modulates various clinical parameters within normal ranges for a healthy person.


Subject(s)
Andrographis , Blood Coagulation/drug effects , Blood Pressure/drug effects , Plant Preparations/pharmacology , Administration, Oral , Adult , Blood Chemical Analysis , Capsules , Female , Humans , Male , Phytotherapy , Plant Preparations/administration & dosage , Pulse , Thailand
3.
J Appl Toxicol ; 36(6): 790-801, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26234195

ABSTRACT

Perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) are the two most popular surfactants among perfluorinated compounds (PFCs), with a wide range of uses. Growing evidence suggests that PFCs have the potential to interfere with estrogen homeostasis, posing a risk of endocrine-disrupting effects. This in vitro study aimed to investigate the estrogenic effect of these compounds on T47D hormone-dependent breast cancer cells. PFOS and PFOA (10(-12) to 10(-4) M) were not able to induce estrogen response element (ERE) activation in the ERE luciferase reporter assay. The ERE activation was induced when the cells were co-incubated with PFOS (10(-10) to 10(-7) M) or PFOA (10(-9) to 10(-7) M) and 1 nM of 17ß-estradiol (E2). PFOS and PFOA did not modulate the expression of estrogen-responsive genes, including progesterone (PR) and trefoil factor (pS2), but these compounds enhanced the effect of E2-induced pS2 gene expression. Neither PFOS nor PFOA affected T47D cell viability at any of the tested concentrations. In contrast, co-exposure with PFOS or PFOA and E2 resulted in an increase of E2-induced cell viability, but no effect was found with 10 ng ml(-1) EGF co-exposure. Both compounds also intensified E2-dependent growth in the proliferation assay. ERK1/2 phosphorylation was increased by co-exposure with PFOS or PFOA and E2, but not with EGF. Collectively, this study shows that PFOS and PFOA did not possess estrogenic activity, but they enhanced the effects of E2 on estrogen-responsive gene expression, ERK1/2 activation and the growth of the hormone-deprived T47D cells. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Alkanesulfonic Acids/toxicity , Breast Neoplasms/chemically induced , Caprylates/toxicity , Endocrine Disruptors/toxicity , Estradiol/agonists , Estrogens/agonists , Fluorocarbons/toxicity , Surface-Active Agents/toxicity , Alkanesulfonic Acids/antagonists & inhibitors , Butadienes/pharmacology , Caprylates/antagonists & inhibitors , Carcinogens, Environmental/chemistry , Carcinogens, Environmental/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Endocrine Disruptors/chemistry , Estradiol/pharmacology , Estrogens/pharmacology , Female , Fluorocarbons/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter/drug effects , Humans , MAP Kinase Signaling System/drug effects , Neoplasm Proteins/agonists , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nitriles/pharmacology , Osmolar Concentration , Protein Kinase Inhibitors/pharmacology , Response Elements/drug effects , Surface-Active Agents/chemistry , Trefoil Factor-1/agonists , Trefoil Factor-1/genetics , Trefoil Factor-1/metabolism
4.
Environ Toxicol ; 31(9): 1133-46, 2016 Sep.
Article in English | MEDLINE | ID: mdl-25728338

ABSTRACT

Arsenic (As) is considered a major environmental health threat worldwide due to its widespread contamination in drinking water. Recent studies reported that arsenic is a potential xenoestrogen as it interfered with the action of estrogen (E2) and estrogen receptor (ER) signaling. The present study investigated the effects of sodium arsenite (NaAsO2 ) on estrogen signaling in human breast cancer cells. The results demonstrated that NaAsO2 dose-dependently increased viability of hormone-dependent breast cancer MCF-7 and T47D cells expressing both ERα and ERß but not hormone-independent MDA-MB-231 cells expressing ERß. These suggested ERα contribution to NaAsO2 -stimulated breast cancer cells growth. NaAsO2 induced down-regulation of ERα but up-regulation of ERß protein expressions in T47D cells. Moreover, NaAsO2 dose-dependently inhibited E2-induced ER transcriptional activity as it decreased E2-mediated ERE-luciferase transcription activation and PgR mRNA transcription but increased pS2 mRNA transcription. However, NaAsO2 induced both rapid and sustained activation of ERK1/2 and increased in phosphorylation of ERα at serine 118 residue, c-fos and c-myc protein expressions. These results indicated that NaAsO2 interferes the genomic estrogen-signaling pathway but induces activation of a rapid nongenomic signal transduction through ERK1/2 pathway which may contribute to its proliferative effect on hormone-dependent breast cancer cells. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1133-1146, 2016.


Subject(s)
Arsenites/toxicity , Estrogen Receptor alpha/metabolism , Signal Transduction/drug effects , Sodium Compounds/toxicity , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation/drug effects , Estradiol/pharmacology , Estrogen Receptor beta/metabolism , Female , Humans , MCF-7 Cells , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Presenilin-2/genetics , Presenilin-2/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/metabolism , Transcriptional Activation/drug effects , Up-Regulation/drug effects
5.
Int J Cancer ; 136(6): 1254-62, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25070670

ABSTRACT

It has become axiomatic that critical windows of susceptibility to genotoxins exist and that genetic damage in utero may be a trigger for later life cancers. Data supporting this critical window hypothesis are remarkably few. This study provides a quantitative bridge between DNA damage by the liver carcinogen aflatoxin B1 (AFB1 ) during prenatal development and the risk of later life genetic disease. AFB1 was given to pregnant C57BL/6J mice, carrying F1 gestation day 14 (GD14) embryos of the B6C3F1 genotype. Ultra-high performance liquid chromatography and mass spectrometry (UPLC-MS) using aflatoxin-(15) N5 -guanine adduct standards afforded measurement of the AFB1 -N(7) -Gua and AFB1 -FAPY adducts 6-hr post dosing in liver DNA of mothers and embryos. A parallel cohort gave birth and the livers of the F1 were analyzed for mutations in the gpt gene at 3 and 10 weeks of age. The data revealed mutational spectra dominated by G:C to T:A mutations in both the mother and offspring that are characteristic of AFB1 and distinct from background. It was shown that adducts in GD14 embryos were 20-fold more potent inducers of mutagenesis than adducts in parallel-dosed adults. This sensitivity enhancement correlated with Ki67 staining of the liver, reflecting the proliferative potential of the tissue. Taken together, these data provide insight into the relative genetic risks of prenatal and adult exposures to AFB1 . Early life exposure, especially during the embryonic period, is strikingly more mutagenic than treatment later in life. Moreover the data provide a baseline against which risk prevention strategies can be evaluated.


Subject(s)
Aflatoxin B1/toxicity , Carcinogens/toxicity , Fetus/drug effects , Liver/drug effects , Mutation , Animals , Cell Proliferation/drug effects , DNA Adducts/analysis , Humans , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL
6.
Planta Med ; 80(7): 533-43, 2014 May.
Article in English | MEDLINE | ID: mdl-24782229

ABSTRACT

Andrographis paniculata is an important herbal medicine widely used in several Asian countries for the treatment of various diseases due to its broad range of pharmacological activities. The present study reports that A. paniculata extracts potently inhibit the growth of liver (HepG2 and SK-Hep1) and bile duct (HuCCA-1 and RMCCA-1) cancer cells. A. paniculata extracts with different contents of major diterpenoids, including andrographolide, 14-deoxy-11,12-didehydroandrographolide, neoandrographolide, and 14-deoxyandrographolide, exhibited a different potency of growth inhibition. The ethanolic extract of A. paniculata at the first true leaf stage, which contained a high amount of 14-deoxyandrographolide but a low amount of andrographolide, showed a cytotoxic effect to cancer cells about 4 times higher than the water extract of A. paniculata at the mature leaf stage, which contained a high amount of andrographolide but a low amount of 14-deoxyandrographolide. Andrographolide, not 14-deoxy-11,12-didehydroandrographolide, neoandrographolide, or 14-deoxyandrographolide, possessed potent cytotoxic activity against the growth of liver and bile duct cancer cells. The cytotoxic effect of the water extract of A. paniculata at the mature leaf stage could be explained by the present amount of andrographolide, while the cytotoxic effect of the ethanolic extract of A. paniculata at the first true leaf stage could not. HuCCA-1 cells showed more sensitivity to A. paniculata extracts and andrographolide than RMCCA-1 cells. Furthermore, the ethanolic extract of A. paniculata at the first true leaf stage increased cell cycle arrest at the G0/G1 and G2/M phases, and induced apoptosis in both HuCCA-1 and RMCCA-1 cells. The expressions of cyclin-D1, Bcl-2, and the inactive proenzyme form of caspase-3 were reduced by the ethanolic extract of A. paniculata in the first true leaf stage treatment, while a proapoptotic protein Bax was increased. The cleavage of poly (ADP-ribose) polymerase was also found in the ethanolic extract of A. paniculata in the first true leaf stage treatment. This study suggests that A. paniculata could be a promising herbal plant for the alternative treatment of intrahepatic cholangiocarcinoma.


Subject(s)
Andrographis/chemistry , Bile Duct Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , Diterpenes/therapeutic use , Phytotherapy , Plant Extracts/therapeutic use , Apoptosis/drug effects , Bile Ducts, Intrahepatic , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Diterpenes/chemistry , Diterpenes/isolation & purification , Humans , Inhibitory Concentration 50 , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plants, Medicinal
7.
Environ Toxicol ; 29(6): 672-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-22859221

ABSTRACT

Arsenic is a widespread contaminant in the environment especially in drinking water. Although it is a known carcinogen in human, the mechanism by which arsenic induces carcinogenesis is not well understood. Among several effects of arsenic, it has been suggested that arsenic-induced vascular endothelial growth factor (VEGF) expression plays a critical role in arsenic carcinogenesis. In the present study, we demonstrated that arsenite induced VEGF expression in neuroblastoma SH-SY5Y cells without induction of HIF-1α, a well-known transcriptional activator for VEGF suggesting that arsenite-induced VEGF expression in SH-SY5Y cells may not require HIF-1α activation. It has been reported that VEGF expression is regulated by multiple transcription factors including ß-catenin. We therefore investigated whether ß-catenin was involved in arsenite-induced VEGF expression in SH-SY5Y cells. Treatment of arsenite caused ß-catenin accumulation in the nucleus. Additionally, arsenite treatment decreased the activity of GSK3, an enzyme that phosphorylates and targets ß-catenin for degradation by proteasome, without activation of its upstream kinase, Akt. Inhibition of PI3K/Akt which negatively regulates GSK3 activity by LY294002 resulted in a decrease in arsenite-mediated ß-catenin nuclear accumulation, and VEGF expression. These results suggested that ß-catenin plays a role in arsenite-induced VEGF in SH-SY5Y cells, and the induction of ß-catenin by arsenite is mediated by inhibition of GSK3 without activating its upstream kinase Akt.


Subject(s)
Arsenites/toxicity , Neuroblastoma/metabolism , Vascular Endothelial Growth Factor A/metabolism , beta Catenin/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Chromones/pharmacology , Glycogen Synthase Kinase 3/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factors/metabolism
8.
Cell Mol Biol Lett ; 18(1): 58-74, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23161404

ABSTRACT

Glycogen synthase kinase-3 (GSK3) and p53 play crucial roles in the mitochondrial apoptotic pathway and are known to interact in the nucleus. However, it is not known if GSK3 has a regulatory role in the mitochondrial translocation of p53 that participates in apoptotic signaling following DNA damage. In this study, we demonstrated that lithium and SB216763, which are pharmacological inhibitors of GSK3, attenuated p53 accumulation and caspase-3 activation, as shown by PARP cleavage induced by the DNA-damaging agents doxorubicin, etoposide and camptothecin. Furthermore, each of these agents induced translocation of p53 to the mitochondria and activated the mitochondrial pathway of apoptosis, as evidenced by the release of cytochrome C from the mitochondria. Both mitochondrial translocation of p53 and mitochondrial release of cytochrome C were attenuated by inhibition of GSK3, indicating that GSK3 promotes the DNA damage-induced mitochondrial translocation of p53 and the mitochondrial apoptosis pathway. Interestingly, the regulation of p53 mitochondrial translocation by GSK3 was only evident with wild-type p53, not with mutated p53. GSK3 inhibition also reduced the phosphorylation of wild-type p53 at serine 33, which is induced by doxorubicin, etoposide and camptothecin in the mitochondria. Moreover, inhibition of GSK3 reduced etoposide-induced association of p53 with Bcl2 and Bax oligomerization. These findings show that GSK3 promotes the mitochondrial translocation of p53, enabling its interaction with Bcl2 to allow Bax oligomerization and the subsequent release of cytochrome C. This leads to caspase activation in the mitochondrial pathway of intrinsic apoptotic signaling.


Subject(s)
Gene Expression Regulation/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Mitochondria/drug effects , Neuroblastoma/genetics , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein/genetics , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Camptothecin/pharmacology , Caspase 3/genetics , Caspase 3/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , DNA Damage , Doxorubicin/pharmacology , Etoposide/pharmacology , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , Mitochondria/genetics , Mitochondria/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Protein Multimerization/drug effects , Protein Transport , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism , bcl-Associated Death Protein/genetics , bcl-Associated Death Protein/metabolism
9.
Chem Res Toxicol ; 25(10): 2194-202, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22971010

ABSTRACT

Hypoxia-inducible factor-1α (HIF-1α) is a critical regulator of cellular responses to hypoxia. Under normoxic conditions, the cellular HIF-1α level is regulated by hydroxylation by prolyl hydroxylases (PHDs), ubiquitylation, and proteasomal degradation. During hypoxia, degradation decreases, and its intracellular level is increased. Exogenously administered nitric oxide (NO)-donor drugs stabilize HIF-1α; thus, NO is suggested to mimic hypoxia. However, the role of low levels of endogenously produced NO generated during hypoxia in HIF-1α stabilization has not been defined. Here, we demonstrate that NO and reactive oxygen species (ROS) produced endogenously by human colon carcinoma HCT116 cells are responsible for HIF-1α accumulation in hypoxia. The antioxidant N-acetyl-L-cysteine (NAC) and NO synthase inhibitor N(G)-monomethyl L-arginine (L-NMMA) effectively reduced HIF-1α stabilization and decreased HIF-1α hydroxylation. These effects suggested that endogenous NO and ROS impaired PHD activity, which was confirmed by reversal of L-NMMA- and NAC-mediated effects in the presence of dimethyloxaloylglycine, a PHD inhibitor. Thiol reduction with dithiothreitol decreased HIF-1α stabilization in hypoxic cells, while dinitrochlorobenzene, which stabilizes S-nitrosothiols, favored its accumulation. This suggested that ROS- and NO-mediated HIF-1α stabilization involved S-nitrosation, which was confirmed by demonstrating increased S-nitrosation of PHD2 during hypoxia. Our results support a regulatory mechanism of HIF-1α during hypoxia in which endogenously generated NO and ROS promote inhibition of PHD2 activity, probably by its S-nitrosation.


Subject(s)
Colonic Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Cell Hypoxia , Colon/cytology , Colon/metabolism , Colon/pathology , Colonic Neoplasms/pathology , HCT116 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Hypoxia-Inducible Factor-Proline Dioxygenases , Nitrosation , Procollagen-Proline Dioxygenase/metabolism
10.
Proc Natl Acad Sci U S A ; 106(34): 14547-51, 2009 Aug 25.
Article in English | MEDLINE | ID: mdl-19706542

ABSTRACT

The transcription factor NF-E2-related nuclear factor 2 (Nrf2) regulates expression of genes that protect cells from oxidative damage. Here, we characterized nitric oxide (*NO)-induced Nrf2-Kelch-like ECH-associated protein 1 (Keap1) signaling and its role in counteracting *NO-induced apoptosis of human colon cancer HCT116 cells. Nrf2 was localized in the cytoplasm in control cells; *NO triggered its rapid nuclear accumulation, transcriptional activation, and up-regulation of HO-1, NQO1, and GCL, but not GST A4 and P1 subunits. Nrf2 accumulation in the nucleus was also associated with enhanced transcription and posttranscriptional modifications. (S)-nitrosation of Keap1 may contribute to nuclear accumulation of Nrf2 by facilitating its dissociation from Keap1, thus initiating *NO-mediated Nrf2-Keap1 signaling. *NO-mediated induction of ARE-dependent genes occurred well before apoptosis, as judged by caspase 3 activation. Collectively, these results show that the Nrf2-Keap1 signaling pathway mediates protective cellular responses to mitigate *NO-induced damage and may contribute to the relative resistance of HCT116 to *NO-induced cytotoxicity.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Nitric Oxide/pharmacology , Signal Transduction/drug effects , Apoptosis/drug effects , Blotting, Western , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cytoplasm/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Kelch-Like ECH-Associated Protein 1 , Microscopy, Confocal , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/genetics , Nitrosation , Protein Transport/drug effects , Reverse Transcriptase Polymerase Chain Reaction
11.
J Appl Toxicol ; 30(7): 649-55, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20981856

ABSTRACT

Acrylonitrile (ACN) is a chemical that is widely used in the production of plastics, acrylic fibers, synthetic rubbers and resins. It has been reported that ACN can cause oxidative stress, a condition which is well recognized as an apoptotic initiator; however, information regarding ACN-induced apoptosis is limited. This present study investigated whether ACN induces apoptosis in human neuroblastoma SH-SY5Y cells, and whether its apoptotic induction involves oxidative stress. The results showed that ACN caused activation of caspase-3, a key enzyme involved in apoptosis, in a dose- and time-dependent manner. Detection of sub-G1 apoptotic cell death and apoptotic nuclear condensation revealed that ACN caused an increase in the number of apoptotic cells indicating ACN induces apoptosis in SH-SY5Y cells. ACN dose- and time-dependently increased the level of proapoptotic protein, Bax. Pretreatment with N-acetylcysteine (NAC), an antioxidant, attenuated caspase-3 activation by ACN, as evidenced by a reduction in proteolysis of PARP, a known caspase-3 substrate, as well as in the number of sub-G1 apoptotic cells. Moreover, induction of Bax by ACN was abolished by NAC. Taken together, the results indicate that ACN induces apoptosis in SH-SY5Y cells via a mechanism involving generation of oxidative stress-mediated Bax induction.


Subject(s)
Acrylonitrile/toxicity , Apoptosis/physiology , Oxidative Stress/physiology , Acetylcysteine/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Caspase 3/analysis , Caspase 3/metabolism , Cell Death/physiology , Cell Line, Tumor , Cell Survival/physiology , Enzyme Activation , Humans , Neuroblastoma , Oxidative Stress/drug effects , Poly(ADP-ribose) Polymerases/metabolism , bcl-2-Associated X Protein/biosynthesis , bcl-2-Associated X Protein/metabolism
12.
Toxicol Rep ; 6: 426-430, 2019.
Article in English | MEDLINE | ID: mdl-31193040

ABSTRACT

Andrographis paniculata is widely used in traditional herbal medicines for the treatment of common cold, fever and diarrhea, in many regions of Scandinavia and Asia, including Thailand. The pharmacological activities of A. paniculata are mainly attributed to active diterpenoids including 14-deoxyandrographolide, which is uniquely high in first true leaf ethanolic extract (FTLEE) of A. paniculata. In this study, the acute toxicity of the standardized FTLEE of A. paniculata was examined according to the OECD test guideline No. 420. Mice were divided into four groups of each sex and orally received the standardized FTLEE of A. paniculata (0, 300, 2000, or 5000 mg/kg BW). Post-treatment, body weight, signs of toxicity, and/or mortality were observed for 14 days. At Day 15, animals were euthanized, internal organs were observed grossly, and blood samples collected were subjected to hematology and clinical biochemistry analyses. The results showed that all treated animals survived and no apparent adverse effects were observed during the duration of the study. Gross necropsy observation revealed no lesion in any organ of all the standardized FTLEE-treated mice. Although significant alterations in BUN, lymphocytes, neutrophils, hematocrit and hemoglobin were observed, these alterations were not treatment-related toxic effects. Therefore, we concluded that a single oral administration of the standardized FTLEE of A. paniculata with an upper fixed dose of 5000 mg/kg BW has no significant acute toxicological effects.

13.
J Toxicol Environ Health A ; 71(16): 1109-23, 2008.
Article in English | MEDLINE | ID: mdl-18569623

ABSTRACT

Methylmercury (MeHg) is an environmental toxicant that is known to induce lymphocyte apoptosis; however, little is known about the molecular mechanism involved. Data showed that MOLT-3 cells were more sensitive to MeHg-induced cytotoxic effects than Jurkat clone E6-1 cells, suggesting that the lymphocytic muscarinic cholinergic system may be involved since the expressions of five subtypes (M1-M5) of muscarinic acetylcholine receptor (mAChR) in MOLT-3 cells are higher than in Jurkat cells. The role of mAChR-linked pathways in MeHg-induced apoptosis in human leukemic T cells was examined in this study. Treatment of the MOLT-3 cells with 1 microM MeHg produced induction of c-Fos expression, apoptotic cell death, and downregulation of mAChR. MeHg-induced c-Fos expression was significantly reduced by pretreatment with atropine (a nonselective mAChR antagonist), or 4-DAMP (a selective M1/M3 mAChR antagonist), whereas pirenzipine (a selective M1 mAChR antagonist) or himbazine (a selective M2/M4 mAChR antagonist) did not reduce this induction, suggesting that MeHg-induced c-Fos expression through the activation of the mAChR, at least M3 subtype, is involved. Pretreatment with 4-DAMP or SB 203580 (a specific p38 inhibitor) resulted in decreases in the level of phosphorylated p38, c-Fos expression, and apoptotic cell death induced by MeHg. Taken together, these data suggest that the mAChR-p38-dependent pathway participates in the increase of c-Fos expression, which is involved in MeHg-induced lymphocyte apoptosis. In addition, a noncytotoxic concentration of MeHg (0.1 microM) inhibited PHA/PMA-stimulated interleukin (IL)-2 production, and this inhibition was reversed by pretreatment with atropine or 4-DAMP. Overall, this study provides initial evidence that MeHg may alter the immune system by targeting the lymphocytic mAChR.


Subject(s)
Apoptosis/drug effects , Genes, fos/drug effects , Methylmercury Compounds/toxicity , Receptors, Muscarinic/drug effects , T-Lymphocytes/drug effects , Drug Interactions , Humans , Jurkat Cells , Muscarinic Antagonists/pharmacology , Piperidines/pharmacology , Receptors, Muscarinic/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism , Tumor Cells, Cultured
14.
Int J Oncol ; 53(1): 177-188, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29693152

ABSTRACT

Cholangiocarcinoma (CCA) is a malignant tumor of the biliary epithelium associated with Opisthorchis viverrini, primary sclerosing cholangitis and hepatitis viral infection. In the global population, men have higher incidence rates for CCA than women; thus, a gender disparity in the progression of chronic inflammation of the biliary duct leading to malignancy may involve the effects of estrogen (E2). Genistein (GE), a prominent phytoestrogen found in soy products, is an estrogen receptor ß (ERß) agonist and a tyrosine kinase inhibitor. The present study investigated the effects of GE on the growth of CCA cells by cell viability assay. The effects on signaling proteins were detected by western blot analysis and ELISA. Gene expression was examined by RT-qPCR. Two human intrahepatic CCA cell lines, HuCCA­1 and RMCCA­1, were utilized. GE (50­200 µM) reduced the viability of the two cell lines, and also inhibited the activation of epidermal growth factor receptor (EGFR) and AKT, as evidenced by decreasing protein levels of phosphorylated (p)-EGFR (Tyr1173) and p­AKT (Ser473), respectively. GE altered the mitogen­activated protein kinase signaling cascade by mediating decreased protein levels of p­p38 and increased protein levels of p­ERK1/2. GE significantly decreased the levels of interleukin 6 (IL6) and induced the expression of inducible nitric oxide synthase (iNOS). GE also downregulated the expression of p­ERα (Ser118) protein and ERα mRNA levels. Finally, GE induced the downregulation of the protein levels of ERß. Of note, E2 deprivation potentiated the GE-induced reduction of p­EGFR (Tyr1173) and total AKT proteins and production of IL6, and mediated the downregulation of GE-induced iNOS protein. In conclusion, GE inhibited the growth of human CCA cell lines by reducing the activation of EGFR and AKT, and by attenuating the production of IL6. E2 and ER were also involved in the growth-inhibitory effect of GE in CCA cells.


Subject(s)
Cholangiocarcinoma/drug therapy , ErbB Receptors/genetics , Genistein/administration & dosage , Interleukin-6/genetics , Oncogene Protein v-akt/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , ErbB Receptors/antagonists & inhibitors , Estrogen Receptor beta/agonists , Estrogen Receptor beta/genetics , Estrogens/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Oncogene Protein v-akt/antagonists & inhibitors , Phosphorylation , Signal Transduction/drug effects
15.
Food Chem Toxicol ; 118: 595-607, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29890199

ABSTRACT

Previous studies showed that glyphosate stimulates breast cancer cell growth via estrogen receptors. The present study investigated the effect of glyphosate on the estrogen signaling pathway involved in the induction of cholangiocarcinoma (CCA) cell growth. HuCCA-1, RMCCA-1 and MMNK-1 were chosen for comparison. The effects of glyphosate on cell growth, cell cycle and molecular signaling pathways were measured. The results showed that HuCCA-1 cells expressed estrogen receptor alpha (ERα), while ERα was not detected in RMCCA-1 and MMNK-1 cells. ERα was mostly expressed in cytoplasmic compartment of HuCCA-1 cells. Estradiol (E2) (10-11-10-5 M) induced cell proliferation in HuCCA-1 but not in RMCCA-1 and MMNK-1 cells. Glyphosate at the same concentration range also induced HuCCA-1 cell proliferation. The S phase of the cell cycle, and protein levels of the cyclin family were significantly increased after treatment of glyphosate or E2. Both compounds also induced the expression of proliferative signaling-related proteins including ERα, VEGFR2, pERK, PI3K(p85), and PCNA. These effects of glyphosate and E2 were abolished by the ER antagonist, 4-hydroxytamoxifen and U0126, a MEK inhibitor. The data from this study indicate that glyphosate can induce cell growth in ERα positive CCA cells through non-genomic estrogen receptor/ERK1/2 signaling pathway.


Subject(s)
Cholangiocarcinoma/pathology , Estrogen Receptor alpha/drug effects , Glycine/analogs & derivatives , Herbicides/toxicity , MAP Kinase Signaling System/drug effects , Cell Line , Cell Line, Tumor , Cholangiocarcinoma/metabolism , Estrogen Receptor alpha/metabolism , Glycine/toxicity , Humans , Signal Transduction/drug effects , Glyphosate
16.
Cancer Res ; 63(22): 8037-50, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14633737

ABSTRACT

Aromatase inhibitors have recently been reported to be more effective than the antiestrogen tamoxifen (Tam) in treating breast cancer. Here, we studied the mechanisms and signaling pathways of cell growth, cell cycle progression, and apoptosis induced by three aromatase inhibitors: letrozole (Let), anastrozole, and 4-hydroxyandrostenedione in comparison with estrogen withdrawal (E2W) and antiestrogens Tam and faslodex. Estrogen-dependent human breast cancer cells stably transfected with aromatase (MCF-7Ca) were used. All treatments induced growth suppression and cell cycle arrest at the G(0)-G(1) phase that was associated with up-regulation of p53 and p21 protein and mRNA levels and down-regulation of cyclin D1 and c-myc mRNA. The apoptotic index was increased 4-7 fold, Bcl-2 protein expression decreased, Bax increased, and caspase-9, caspase-6, and caspase-7 were activated but not caspase-3 and caspase-8. Let and E2W caused regression of tumors of MCF-7Ca cells grown in nude mice and increased the number of cells undergoing apoptosis. In contrast, Tam and faslodex did not induce tumor regression and a lower number of apoptotic cells was detected. Cleavage of poly(ADP-ribose) polymerase was detected. Treatment with Let, Tam, or E2W resulted in a dose- and time-dependent increase in active caspase-7 and up-regulation of p53 and p21 protein. Although the mechanisms involved appeared to be similar for antiestrogens and aromatase inhibitors, the most significant effects occurred with Let, which were significantly greater than with E2W and consistent with marked effects of Let on tumor and cell growth.


Subject(s)
Apoptosis/drug effects , Aromatase Inhibitors , Breast Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Estrogen Receptor Modulators/pharmacology , Animals , Apoptosis/genetics , Apoptosis/physiology , Aromatase/genetics , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Caspases/metabolism , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , Down-Regulation/drug effects , Enzyme Activation/drug effects , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Ovariectomy , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction/drug effects , Transfection , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Up-Regulation/drug effects , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein
17.
J Ethnopharmacol ; 179: 156-61, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26721218

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Flower, seed, and fruit of longan (Dimocarpus longan Lour.) have been used in the Traditional Chinese Medicine (TCM) serving as a common herb in relief of swelling which can be applied in cases of inflammatory diseases. However, the scientific evidence related to their effects on inflammation especially the possible cellular and molecular mechanisms of longan need to be clarified. AIM OF THE STUDY: To evaluate the anti-inflammatory effect of the various parts of longan including flower, seed, and pulp. The mechanisms and molecular targets involved in their effects were also investigated. MATERIALS AND METHODS: Different longan extracts were analyzed for their bioactive compounds and evaluated for anti-inflammation. Corilagin, ellagic acid, and gallic acid were detected using HPLC-DAD. In vitro anti-inflammatory effect of longan extracts and their polysaccharides were examined by analyzing nitric oxide (NO) production in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. Expression of the proteins that involved in NO production was detected by Western blot. RESULTS: Flower extract contained the highest amounts of total phenolics, total flavonoids, proanthocyanidins, corilagin and ellagic acid when compared to seed and pulp extracts. The extracts of different longan parts inhibited LPS-induced NO production with different potency. The highest potency for the inhibition of NO production was shown with flower extract follow by seed and pulp (IC50=128.2, 1127.4, and 1260.2µgmL(-1), respectively). The mechanisms of this anti-NO production were associated with their NO scavenging effect and their decreasing the expression and catalytic activity of an inducible nitric oxide synthase (iNOS). Moreover, these longan extracts suppressed LPS-induced degradation of IκBα and activation of NF-κB, activator protein-1 (AP-1), Akt, and mitogen activated protein kinases (MAPKs). CONCLUSION: These results suggest that the longan extracts possess anti-inflammatory property. Therefore, longan could provide potential dietary supplement for the treatment of inflammatory-related diseases.


Subject(s)
Lipopolysaccharides/antagonists & inhibitors , Macrophages/metabolism , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Sapindaceae/chemistry , Signal Transduction/drug effects , Transcription Factor AP-1/metabolism , Animals , Flowers/chemistry , Fruit/chemistry , Lipopolysaccharides/toxicity , Macrophages/drug effects , Mice , NF-kappa B/drug effects , Nitric Oxide Synthase Type II/metabolism , Plant Extracts/pharmacology , Polysaccharides/chemistry , Polysaccharides/pharmacology , RAW 264.7 Cells , Seeds/chemistry , Transcription Factor AP-1/drug effects
18.
Environ Health Perspect ; 124(2): 170-5, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26115410

ABSTRACT

BACKGROUND: Millions of individuals worldwide, particularly those living in rural and developing areas, are exposed to harmful levels of inorganic arsenic (iAs) in their drinking water. Inorganic As exposure during key developmental periods is associated with a variety of adverse health effects, including those that are evident in adulthood. There is considerable interest in identifying the molecular mechanisms that relate early-life iAs exposure to the development of these latent diseases, particularly in relationship to cancer. OBJECTIVES: This work summarizes research on the molecular mechanisms that underlie the increased risk of cancer development in adulthood that is associated with early-life iAs exposure. DISCUSSION: Epigenetic reprogramming that imparts functional changes in gene expression, the development of cancer stem cells, and immunomodulation are plausible underlying mechanisms by which early-life iAs exposure elicits latent carcinogenic effects. CONCLUSIONS: Evidence is mounting that relates early-life iAs exposure and cancer development later in life. Future research should include animal studies that address mechanistic hypotheses and studies of human populations that integrate early-life exposure, molecular alterations, and latent disease outcomes.


Subject(s)
Arsenic/toxicity , Environmental Exposure , Neoplasms/chemically induced , Water Pollutants, Chemical/toxicity , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Child , Child, Preschool , Disease Models, Animal , Drinking Water/analysis , Gene Expression Regulation/drug effects , Humans , Infant , Infant, Newborn , Middle Aged , Risk Factors , Young Adult
19.
Clin Cancer Res ; 8(7): 2378-88, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12114443

ABSTRACT

PURPOSE: The aromatase inhibitors letrozole and anastrozole have been approvedrecently as first-line treatment options for hormone-dependent advanced breast cancer. Although it is established that a proportion of patients who relapse on first-line tamoxifen therapy show additional responses to aromatase inhibitors, it has not been determined whether tumors that acquire resistance to aromatase inhibitors in the first line remain sensitive to second-line therapy with antiestrogens. The aim of this study was to determine whether aromatase-transfected and hormone-dependent MCF-7Ca human breast cancer cells remain sensitive to antiestrogens after: (a) long-term growth in steroid-depleted medium in vitro; and (b) long-term treatment with the aromatase inhibitor letrozole in vivo. METHODS: In the first approach, a variant of the MCF-7Ca human breast cancer cell line was selected that had acquired the ability to grow in estrogen-depleted medium after 6-8 months of culture. Steroid-deprived UMB-1Ca cells were analyzed for aromatase activity levels, hormone receptor levels, and sensitivity to estrogens and antiestrogens in vitro and in vivo. In the second approach, established MCF-7Ca breast tumor xenografts were treated with letrozole 10 microg/day for 12 weeks followed by 100 microg/day for 25 weeks until tumors acquired the ability to proliferate in the presence of the drug. Long-term letrozole-treated tumors were then transplanted into new mice, and the effects of antiestrogens and aromatase inhibitors on tumor growth were determined. RESULTS: Steroid-deprived UMB-1Ca breast cancer cells continued to express aromatase activity at levels comparable with the parental cell line. However, compared with MCF-7Ca cells, UMB-1Ca cells expressed elevated levels of functionally active estrogen receptor. The growth of UMB-1Ca cells in vitro was inhibited by the antiestrogens tamoxifen and faslodex and tumor growth in vivo was inhibited by tamoxifen. In the second approach, the time for MCF-7Ca tumor xenografts to approximately double in volume after being treated sequentially with the increasing doses of letrozole was thirty-seven weeks. Long-term letrozole-treated tumors continued to express functionally active aromatase. When transplanted into new mice, growth of the long-term letrozole-treated tumors was slowed by tamoxifen and inhibited more effectively by faslodex. Tumor growth was refractory to the aromatase inhibitors anastrozole and formestane but, surprisingly, showed sensitivity to letrozole. CONCLUSIONS: Steroid-deprived UMB-1Ca human breast cancer cells selected in vitro and long-term letrozole-treated MCF-7Ca breast tumor xenografts remain sensitive to second-line therapy with antiestrogens and, in particular, to faslodex. This finding is associated with increased expression of functionally active estrogen receptor after steroid-deprivation of MCF-7Ca human breast cancer cells in vitro.


Subject(s)
Aromatase Inhibitors , Enzyme Inhibitors/therapeutic use , Estradiol/analogs & derivatives , Estrogen Antagonists/therapeutic use , Mammary Neoplasms, Experimental/pathology , Neoplasms, Hormone-Dependent/drug therapy , Nitriles/therapeutic use , Triazoles/therapeutic use , Animals , Aromatase/genetics , Disease Models, Animal , Estradiol/metabolism , Estradiol/therapeutic use , Female , Fulvestrant , Humans , In Vitro Techniques , Letrozole , Mammary Neoplasms, Experimental/enzymology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Hormone-Dependent/pathology , Receptors, Estrogen/metabolism , Tamoxifen/therapeutic use , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
20.
Int J Oncol ; 46(6): 2317-26, 2015.
Article in English | MEDLINE | ID: mdl-25815516

ABSTRACT

Cholangiocarcinoma (CCA) is a malignant cancer of the biliary tract and its occurrence is associated with chronic cholestasis which causes an elevation of bile acids in the liver and bile duct. The present study aimed to investigate the role and mechanistic effect of bile acids on the CCA cell growth. Intrahepatic CCA cell lines, RMCCA-1 and HuCCA-1, were treated with bile acids and their metabolites to determine the growth promoting effect. Cell viability, cell cycle analysis, EdU incorporation assays were conducted. Intracellular signaling proteins were detected by western immunoblotting. Among eleven forms of bile acids and their metabolites, only taurolithocholic acid (TLCA) concentration dependently (1-40 µM) increased the cell viability of RMCCA-1, but not HuCCA-1 cells. The cell cycle analysis showed induction of cells in the S phase and the EdU incorporation assay revealed induction of DNA synthesis in the TLCA-treated RMCCA-1 cells. Moreover, TLCA increased the phosphorylation of EGFR, ERK 1/2 and also increased the expression of cyclin D1 in RMCCA-1 cells. Furthermore, TLCA-induced RMCCA-1 cell growth could be inhibited by atropine, a non-selective muscarinic acetylcholine receptor (mAChR) antagonist, AG 1478, a specific EGFR inhibitor, or U 0126, a specific MEK 1/2 inhibitor. These results suggest that TLCA induces CCA cell growth via mAChR and EGFR/EKR1/2 signaling pathway. Moreover, the functional presence of cholinergic system plays a certain role in TLCA-induced CCA cell growth.


Subject(s)
Bile Acids and Salts/adverse effects , Bile Duct Neoplasms/pathology , Cholangiocarcinoma/pathology , MAP Kinase Signaling System/drug effects , Taurolithocholic Acid/adverse effects , Bile Acids and Salts/pharmacology , Bile Duct Neoplasms/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cholangiocarcinoma/metabolism , Cyclin D1/metabolism , ErbB Receptors/metabolism , Humans , Phosphorylation , Receptors, Muscarinic , Taurolithocholic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL