Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Biochem Pharmacol ; 198: 114973, 2022 04.
Article in English | MEDLINE | ID: mdl-35189109

ABSTRACT

Arsenite, a well-established human carcinogen and toxic compound, promotes the formation of mitochondrial superoxide (mitoO2-) via a Ca2+-dependent mechanism, in which an initial stimulation of the inositol 1, 4, 5-trisphosphate receptor (IP3R) is followed by the activation of the ryanodine receptor (RyR), critical for providing Ca2+ to the mitochondria. We now report that, under the same conditions, arsenite triggers endoplasmic reticulum (ER) stress and a threefold increase in ER oxidoreductin 1α (ERO1 α) levels in proliferating U937 cells. EN460, an inhibitor of ERO1 α, recapitulated all the effects associated with RyR inhibition or downregulation, including prevention of RyR-induced Ca2+ accumulation in mitochondria and the resulting O2-. formation. Quantitatively similar results were obtained in inhibitor studies performed in terminally differentiated wild type C2C12 cells. Moreover, ERO1 α knockout C2C12 myotubes responded to arsenite as their wild type counterpart supplemented with EN460. As a final note, arsenite enhanced the expression of ERO1 α via a mechanism mediated by Ca2+ release from both the IP3R and RyR. We therefore conclude that arsenite activates a positive feedback amplification cycle between Ca2+ levels and ERO1 α in the ER, by which IP3R-dependent Ca2+ induces ERO1 α and ERO1 α promotes Ca2+ release via RyR, thereby amplifying the initial Ca2+ load and causing the mitochondrial accumulation of the cation, critical for mitoO2- formation.


Subject(s)
Calcium Signaling , Membrane Glycoproteins , Oxidoreductases , Ryanodine Receptor Calcium Release Channel , Arsenites/adverse effects , Calcium/metabolism , Humans , Membrane Glycoproteins/metabolism , Mitochondria/metabolism , Oxidoreductases/metabolism , Reactive Oxygen Species/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , U937 Cells
2.
Oxid Med Cell Longev ; 2022: 9574473, 2022.
Article in English | MEDLINE | ID: mdl-35069981

ABSTRACT

Chronic exposure to inorganic arsenic is a major environmental public health issue worldwide affecting more than 220 million of people. Previous studies have shown the correlation between arsenic poisoning and cellular senescence; however, knowledge regarding the mechanism and effective prevention measures has not been fully studied. First, the associations among the ERK/CEBPB signaling pathway, oxidative stress, and arsenic-induced skin cell senescence were confirmed using the HaCaT cell model. In the arsenic-exposed group, the relative mRNA and protein expressions of ERK/CEBPB signaling pathway indicators (ERK1, ERK2, and CEBPB), cell cycle-related genes (p21, p16INK4a), and the secretion of SASP (IL-1α, IL-6, IL-8, TGF-ß1, MMP-1, MMP-3, EGF, and VEGF) and the lipid peroxidation product (MDA) were significantly increased in cells (P < 0.05), while the activity of antioxidant enzyme (SOD, GSH-Px, and CAT) was significantly decreased (P < 0.05), and an increased number of cells accumulated in the G1 phase (P < 0.05). Further Kaji-ichigoside F1 intervention experiments showed that compared to that in the arsenic-exposed group, the expression level of the activity of antioxidant enzyme was significantly increased in the Kaji-ichigoside F1 intervention group (P < 0.05), but the indicators of ERK/CEBPB signaling pathway, cell cycle-related genes, and SASP were significantly decreased (P < 0.05), and the cell cycle arrest relieved to a certain extent (P < 0.05). Our study provides some limited evidence that the ERK/CEBPB signaling pathway is involved in low-dose arsenic-induced skin cell senescence, through regulating oxidative stress. The second major finding was that Kaji-ichigoside F1 can downregulate the ERK/CEBPB signaling pathway and regulate the balance between oxidation and antioxidation, alleviating arsenic-induced skin cell senescence. This study provides experimental evidence for further understanding of Kaji-ichigoside F1, a natural medicinal plant that may be more effective in preventing and controlling arsenic poisoning.


Subject(s)
Arsenites/adverse effects , Cellular Senescence/drug effects , Drugs, Chinese Herbal/therapeutic use , Skin/drug effects , Drugs, Chinese Herbal/pharmacology , Humans
3.
Sci Rep ; 11(1): 22121, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34764389

ABSTRACT

Arsenic exposure poses numerous threats to human health. Our previous work in mice has shown that arsenic causes anemia by inhibiting erythropoiesis. However, the impacts of arsenic exposure on human erythropoiesis remain largely unclear. We report here that low-dose arsenic exposure inhibits the erythroid differentiation of human hematopoietic progenitor cells (HPCs). The impacts of arsenic (in the form of arsenite; As3+) on red blood cell (RBC) development was evaluated using a long-term culture of normal human bone marrow CD34+-HPCs stimulated in vitro to undergo erythropoiesis. Over the time course studied, we analyzed the expression of the cell surface antigens CD34, CD71 and CD235a, which are markers commonly used to monitor the progression of HPCs through the stages of erythropoiesis. Simultaneously, we measured hemoglobin content, which is an important criterion used clinically for diagnosing anemia. As compared to control, low-dose As3+ exposure (100 nM and 500 nM) inhibited the expansion of CD34+-HPCs over the time course investigated; decreased the number of committed erythroid progenitors (BFU-E and CFU-E) and erythroblast differentiation in the subsequent stages; and caused a reduction of hemoglobin content. These findings demonstrate that low-dose arsenic exposure impairs human erythropoiesis, likely by combined effects on various stages of RBC formation.


Subject(s)
Antigens, CD34/metabolism , Arsenites/adverse effects , Cell Differentiation/drug effects , Erythroid Precursor Cells/drug effects , Hematopoietic Stem Cells/drug effects , Hemoglobins/metabolism , Anemia/chemically induced , Anemia/metabolism , Antigens, CD/metabolism , Cells, Cultured , Erythroblasts/drug effects , Erythroblasts/metabolism , Erythroid Precursor Cells/metabolism , Erythropoiesis/drug effects , Glycophorins/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Receptors, Transferrin/metabolism
4.
Toxicol Appl Pharmacol ; 409: 115303, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33141059

ABSTRACT

DNA damage plays a crucial role in the transforming potential of the human carcinogen arsenic. The arsenic biotransformation enzyme AS3MT is known to participate in the generation of ROS after arsenic exposure, whereas MTH1 sanitizes oxidized dNTP pools to prevent the incorporation of damaged bases into DNA. In this work, we sought to assess the role of these two enzymes in the genotoxic and carcinogenic effects of arsenic exposure. Thus, mouse embryonic fibroblasts (MEF), transformed by chronic arsenite exposure, were monitored for DNA damage by the comet and the micronucleus assays at different time-of-exposure intervals lasting for 50 weeks. Results indicate that the oxidative and DNA damage of chronically exposed MEF cells increased time-dependently up to the point of transformation. As3mt expression followed a pattern like that of DNA damage, and its forced inhibition by shRNA technology before transformation resulted in a DNA damage decrease. On the other hand, Mth1 mRNA levels increased after the transformation point, and its forced knock-down increased significantly the levels of DNA damage and decreased the aggressiveness of the oncogenic phenotype. Thus, As3mt and Mth1 have important differential roles in the accumulation of DNA damage linked to the transformation process: while As3mt contributes to the genotoxic effects before the transformation, Mth1 prevents the DNA damage fixation after the acquisition of the oncogenic phenotype. This study demonstrates the influence of As3mt and Mth1 in arsenic DNA damage induction and it is the first to present Mth1 as a candidate modulator biomarker of the tumoral phenotype.


Subject(s)
Arsenic/toxicity , Carcinogenesis/drug effects , Carcinogens/toxicity , Methyltransferases/metabolism , Mutagens/toxicity , Phosphoric Monoester Hydrolases/metabolism , Animals , Arsenites/adverse effects , Carcinogenesis/metabolism , Cell Line , Cells, Cultured , DNA Damage/drug effects , Fibroblasts/drug effects , Mice , Micronucleus Tests/methods , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Phenotype , RNA, Messenger/metabolism
5.
Expert Opin Investig Drugs ; 29(9): 1059-1067, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32735765

ABSTRACT

BACKGROUND: We evaluated the tolerability, pharmacokinetics (PK) and preliminary efficacy of KML001, an oral trivalent arsenical, as a monotherapy in patients with advanced solid tumors. RESEARCH DESIGN AND METHODS: With a standard 3 + 3 design for dose-escalation stage, the planned dose levels of KML001 were 5, 7.5, 10, 12.5, and 15 mg/day for 28 days. Once the maximum tolerated dose was determined, 22 subjects were additionally enrolled for dose-expansion stage. PK analysis was performed in the 5, 10, and 15 mg/day cohort at the dose-escalation stage and also at the dose-expansion stage. Moreover, response was assessed using the standard RECIST 1.1. RESULTS: A total of 45 Korean subjects were enrolled. No DLT was reported at the dose-escalation stage. Three DLTs, two cases of prolonged QTc interval and one of neutropenia, were reported in the 12.5 mg/day cohort at the dose-expansion stage. Higher total daily doses up to 12.5 mg/day of KML001 resulted in higher trough plasma concentrations. Among the 18 subjects who completed 2 cycles of therapy, 15 had progressive disease and 3 had stable disease. CONCLUSIONS: Doses equal to or greater than 10 mg/day KML001 alone were tolerable and produced plasma concentrations higher than biologically relevant targets.


Subject(s)
Antineoplastic Agents/administration & dosage , Arsenites/administration & dosage , Neoplasms/drug therapy , Sodium Compounds/administration & dosage , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Arsenites/adverse effects , Arsenites/pharmacokinetics , Disease Progression , Dose-Response Relationship, Drug , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/pathology , Sodium Compounds/adverse effects , Sodium Compounds/pharmacokinetics , Treatment Outcome
6.
Mol Biol Rep ; 47(2): 1161-1171, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31820315

ABSTRACT

Arsenic is a potent and toxic heavy metal found in the environment that causes health problems, including liver disease, in humans and animals. Chlorogenic acid (CA) is the most abundant caffeoylquinic acid isomer present in plants. This study aims to assess how CA protects the liver tissue following sodium arsenite (NaAsO2)-induced toxicity in mice. Male Swiss mice were allocated into 5 groups: Control, intragastrically administered CA (200 mg/kg), intragastrically administered NaAsO2 (5 mg/kg), and two groups administered with CA (100 and 200 mg/kg) and NaAsO2. CA was administered 30 min before NaAsO2 and all the mice were treated daily for 28 days. To investigate the biochemical, histopathological, immunohistochemical, and molecular changes, blood and liver samples were collected. NaAsO2 treatment increased the liver function biomarkers such as alanine transaminase, aspartate transaminase, alkaline phosphatase, and total bilirubin. Lipid and nitric oxide production was elevated. Glutathione content and the activities of superoxide dismutase, catalase, glutathione peroxidase, and glutathione reductase decreased, indicating a disturbance in redox homeostasis. Histopathological examination revealed a granular degeneration of hepatocytes, infiltration of inflammatory cells, and centrilobular hepatocyte necrosis. Furthermore, tumor necrosis factor-α and interleukin-1ß were upregulated upon NaAsO2 treatment, suggesting the induction of inflammation. Moreover, NaAsO2 triggered apoptosis in the liver by upregulating Bax and caspase-3 and downregulating Bcl-2. However, CA abrogated the biochemical, molecular, and histological changes, reflecting its hepatoprotective role in response to NaAsO2 treatment. Our findings demonstrate that CA could be a potential therapeutic to minimize NaAsO2-induced hepatic injury.


Subject(s)
Apoptosis/drug effects , Arsenites/adverse effects , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Chlorogenic Acid/pharmacology , Oxidative Stress/drug effects , Protective Agents/pharmacology , Sodium Compounds/adverse effects , Animals , Antioxidants/metabolism , Biomarkers , Chemical and Drug Induced Liver Injury/diagnosis , Chemical and Drug Induced Liver Injury/prevention & control , Cytokines/metabolism , Disease Models, Animal , Hepatocytes/drug effects , Hepatocytes/metabolism , Immunohistochemistry , Inflammation Mediators/metabolism , Liver Function Tests , Male , Mice
7.
Int J Mol Sci ; 21(1)2019 Dec 19.
Article in English | MEDLINE | ID: mdl-31861522

ABSTRACT

Telomerase is best known for its function in maintaining telomeres but has also multiple additional, non-canonical functions. One of these functions is the decrease of oxidative stress and DNA damage due to localisation of the telomerase protein TERT into mitochondria under oxidative stress. However, the exact molecular mechanisms behind these protective effects are still not well understood. We had shown previously that overexpression of human telomerase reverse transcriptase (hTERT) in human fibroblasts results in a decrease of mitochondrial DNA (mtDNA) damage after oxidative stress. MtDNA damage caused by oxidative stress is removed via the base excision repair (BER) pathway. Therefore we aimed to analyse whether telomerase is able to improve this pathway. We applied different types of DNA damaging agents such as irradiation, arsenite treatment (NaAsO2) and treatment with hydrogen peroxide (H2O2). Using a PCR-based assay to evaluate mtDNA damage, we demonstrate that overexpression of hTERT in MRC-5 fibroblasts protects mtDNA from H2O2 and NaAsO2 induced damage, compared with their isogenic telomerase-negative counterparts. However, overexpression of hTERT did not seem to increase repair of mtDNA after oxidative stress, but promoted increased levels of manganese superoxide dismutase (MnSOD) and forkhead-box-protein O3 (FoxO3a) proteins during incubation in serum free medium as well as under oxidative stress, while no differences were found in protein levels of catalase. Together, our results suggest that rather than interfering with mitochondrial DNA repair mechanisms, such as BER, telomerase seems to increase antioxidant defence mechanisms to prevent mtDNA damage and to increase cellular resistance to oxidative stress. However, the result has to be reproduced in additional cellular systems in order to generalise our findings.


Subject(s)
Culture Media, Serum-Free/chemistry , Mitochondria/genetics , Superoxide Dismutase/genetics , Telomerase/genetics , Arsenites/adverse effects , Cells, Cultured , DNA Repair , DNA, Mitochondrial/genetics , Forkhead Box Protein O3/metabolism , Humans , Hydrogen Peroxide/adverse effects , Mitochondria/drug effects , Superoxide Dismutase/metabolism , Telomerase/metabolism , Ultraviolet Rays/adverse effects , Up-Regulation
8.
Sci Total Environ ; 686: 1229-1237, 2019 Oct 10.
Article in English | MEDLINE | ID: mdl-31412519

ABSTRACT

Underground drinking water is commonly contaminated with arsenite (As) and fluoride (F) associated with chronic kidney diseases in humans; however, the combined renal toxicity of these pollutants and the underlying mechanisms are still unclear. The aim of the present study was to investigate the interaction between As and F regarding toxic effects on the kidney of rat offspring exposed to pollutants during prenatal and postnatal development. Pregnant rats were randomly divided into four groups that received NaAsO2 (50 mg/L), NaF (100 mg/L), NaAsO2 (50 mg/L) and NaF (100 mg/L) in drinking water, or clean water, respectively, during gestation and lactation. After weaning, six male pups were randomly selected from each group and continued on the same treatment as their mothers for up to three months. The results revealed that subchronic exposure to high-dose As and/or F decreased the organ coefficient of the kidneys and disrupted kidney ultrastructure, moreover inhibited the activity of antioxidant enzymes and increased the generation of malondialdehyde in the kidney. As exposure alone or combined with F led to an upregulation of nuclear factor erythroid 2-related factor-2 (Nrf2) and its regulatory targets (Ho-1, Gclc, and Nqo1), whereas the effect of F alone was not significant. These results suggest that the renal toxicity of As and F is associated with the induction of mitochondrial damage and oxidative stress, and alters the expression of Nrf2 and its regulatory targets. Furthermore, variance analysis results showed that an interaction between As and F in the toxicity process.


Subject(s)
Arsenites/adverse effects , Fluorides/adverse effects , Kidney/drug effects , Oxidative Stress/drug effects , Sexual Maturation/drug effects , Animals , Female , Kidney/ultrastructure , Male , Maternal Exposure/adverse effects , Pregnancy , Random Allocation , Rats , Rats, Sprague-Dawley , Toxicity Tests, Subchronic
9.
Article in English | MEDLINE | ID: mdl-31199764

ABSTRACT

Background Curcumin is extensively used as a therapeutic intervention for treating several ailments. The antioxidant curcumin has an anti-inflammatory and chelating property with arsenic to exhibit a strong therapeutic effect on reproductive organs. This study was undertaken to describe the protective effect of noninvasive administration of curcumin against sodium-arsenite-mediated uterine hazards in female Wistar rats. Methods Twenty-four female Wistar rats were randomly divided into four groups. The treatment was continued for 8 days and given orally sodium arsenite (10 mg/kg body weight) in combination with curcumin (20 mg/kg body weight). Results Our evaluation revealed that 8 days of sodium arsenite (10 mg/kg body weight) treatment reduced the activities of the uterine enzymatic antioxidants superoxide dismutase, catalase, and peroxidase. Blood levels of vitamin B12 and folic acid decreased followed by an increased serum lactate dehydrogenase, homocysteine level, and hepatic metallothionein-1 in arsenic-treated rats. Necrosis of uterine tissue along with the disruption of ovarian steroidogenesis was marked in arsenic-treated rats with an upregulation of uterine NF-κB and IL-6 along with a raised level of serum TNF-α. Oral administration of curcumin (20 mg/kg body weight/day) in arsenic-treated rats significantly reinstated these alterations of the antioxidant system followed by an improvement of ovarian steroidogenesis and the circulating level of B12 and folate along with the downregulation of serum homocysteine, metallothionein-1, and cytokines. Conclusions The findings of this study clearly and strongly elucidated that arsenic-induced oxidative stress in uterus is linked to an alteration of inflammation-signaling biomarkers and these have been protected through the co-administration of curcumin due to its anti-inflammatory, free radical scavenging, and antioxidant activity by the possible regulation of an S-adenosine methionine pool.


Subject(s)
Arsenic/administration & dosage , Curcumin/adverse effects , Cytokines/metabolism , Inflammation/metabolism , Metallothionein/metabolism , Uterus/drug effects , Animals , Antioxidants/pharmacology , Arsenites/adverse effects , Catalase/metabolism , Female , Glutathione/metabolism , Glutathione Peroxidase/metabolism , NF-kappa B/metabolism , Oxidative Stress/drug effects , Peroxidase/metabolism , Rats , Rats, Wistar , Sodium Compounds/adverse effects , Superoxide Dismutase/metabolism , Uterus/metabolism
10.
Cancer Sci ; 110(8): 2629-2642, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31215104

ABSTRACT

Multigenerational adverse effects from the environment such as nutrition and chemicals are among important concerns in environmental health issues. Previously, we have found that arsenite exposure of only F0 females during their pregnancy increases hepatic tumors in the F2 males in C3H mice. In the current study, we investigated the association of DNA methylation with the hepatic tumor increase in the F2 males of the arsenite group. Reduced-representation bisulfite sequencing analysis newly identified that DNA methylation levels of regions around the transcriptional start sites of Tmem54 and Cd74 were decreased and the expression of these genes were significantly increased in the hepatic tumors of F2 males of the arsenite group. The associations between DNA methylation in these regions and gene expression changes were confirmed by treatment of murine hepatoma cell lines and hepatic stellate cell line with 5-aza-2'-deoxycytidine. Overexpression of Cd74 in Hepa1c1c7 cells increased Trib3 expression and suppressed the expression of tumor suppressor genes Id3 and Atoh8. Human database analysis using the Cancer Genome Atlas indicated that TMEM54, CD74, and TRIB3 were significantly increased and that ATOH8 was decreased in hepatocellular carcinoma. The data also showed that high expression of TMEM54 and TRIB3 and low expression of ATOH8 were associated with poor survival. These results suggested that an increase in Tmem54 and Cd74 expression via DNA methylation reduction was involved in the tumor increase in the F2 male offspring by gestational arsenite exposure of F0 females. This study also suggested that genes downstream of Cd74 were involved in tumorigenesis.


Subject(s)
Arsenites/adverse effects , Carcinoma, Hepatocellular/genetics , DNA Methylation/genetics , Liver Neoplasms/genetics , Animals , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Neoplasm/genetics , Carcinoma, Hepatocellular/chemically induced , Cell Line, Tumor , Female , Gene Expression/genetics , Histocompatibility Antigens Class II/genetics , Liver Neoplasms/chemically induced , Male , Mice , Mice, Inbred C3H , Pregnancy
11.
Toxicol Appl Pharmacol ; 378: 114614, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31176655

ABSTRACT

The mechanism of arsenic-induced skin carcinogenesis is not yet fully understood. Chromosomal instability contributes to aneuploidy and is a driving force in carcinogenesis. Arsenic causes mitotic arrest and induces aneuploidy. hsa-miR-186 overexpression is associated with metastatic cancers as well as arsenic-induced squamous cell carcinoma and is reported to target several mitotic regulators. Decreased levels of these proteins can dysregulate chromatid segregation contributing to aneuploidy. This work investigates the potential aneuploidogenic role of hsa-miR-186 in arsenic carcinogenesis. Clones of immortalized human keratinocytes (HaCaT) stably transfected with a hsa-miR-186 expression or empty vector were isolated. Three clones with high and low hsa-miR-186 expression determined by RT-qPCR were selected for further analysis and cultured with 0 or 100 nM NaAsO2 for 8 weeks. Analysis of mitoses revealed that chromosome number and structural abnormalities increased in cells overexpressing hsa-miR-186 and were further increased by arsenite exposure. Double minutes were the dominant structural aberrations. The peak number of chromosomes also increased. Cells with >220 to >270 chromosomes appeared after 2 months in hsa-miR-186 overexpressing cells, indicating multiple rounds of endomitosis had occurred. The fraction of cells with increased chromosome number or structural abnormalities did not increase in passage matched control cells. Levels of selected target proteins were determined by western blot. Expression of BUB1, a predicted hsa-miR-186 target was suppressed in hsa-miR-186 overexpressing clones, but increased with arsenite exposure. CDC27 remained constant under all conditions. These results suggest that overexpression of miR-186 in arsenic exposed tissues likely induces aneuploidy contributing to arsenic-induced carcinogenesis.


Subject(s)
Arsenic/adverse effects , Arsenites/adverse effects , Chromosomal Instability/genetics , Keratinocytes/drug effects , MicroRNAs/genetics , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Line , Humans
12.
Hepatobiliary Pancreat Dis Int ; 18(1): 62-66, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30612929

ABSTRACT

BACKGROUND: Sodium meta-arsenite (NaAsO2, KML001) is a potential oral anticancer agent acting on telomerase and telomere length. This prospective study evaluated its safety, tolerability, and effectiveness as salvage chemotherapy in patients with advanced biliary tract cancer (BTC) resistant to gemcitabine-based chemotherapy. METHODS: Forty-four patients (21 women and 23 men) with advanced BTC and failure history of gemcitabine-based chemotherapy, performance status (PS) 0-2, normal cardiac, hepatic, and renal function were enrolled. Daily dose of KML001 (7.5 mg. p.o.) was administered to eligible subjects for 24 weeks divided into six treatment cycles. Response was evaluated bimonthly using CT. RESULTS: After an average of 1.5 months of treatment (range: 0.5-10.0), 3 patients (6.8%) obtained progression-free status, 23 patients (52.3%) had disease progression, and 18 patients (40.9%) dropped out before evaluation. One patient (2.3%) completed six treatment cycles without progression. During the treatment, morphine dosage kept the same or decreased in 20 patients (47.6%). Nine patients (20.5%) experienced grade-3 adverse events (AEs), while no patient experienced grade-4 AEs. The most common AEs were liver enzyme elevation (11/44, 25%) and anemia (10/44, 22.7%). KML001 was discontinued in six patients (13.6%) due to AEs, including liver toxicity (n = 3), QTc prolongation (n = 2), and abdominal pain (n = 1). CONCLUSIONS: KML001 did not have enough anticancer effect on patients with advanced BTC resistant to gemcitabine. However, KML001 was safe and well-tolerable in terms of AEs and pain control when used as salvage therapy. Further studies are needed to establish arsenic agents as a reliable treatment option in patients with BTC.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arsenites/administration & dosage , Biliary Tract Neoplasms/drug therapy , Salvage Therapy , Sodium Compounds/administration & dosage , Administration, Oral , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Arsenites/adverse effects , Biliary Tract Neoplasms/diagnostic imaging , Biliary Tract Neoplasms/mortality , Biliary Tract Neoplasms/pathology , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Administration Schedule , Drug Resistance, Neoplasm , Female , Humans , Male , Middle Aged , Progression-Free Survival , Prospective Studies , Sodium Compounds/adverse effects , Time Factors , Tomography, X-Ray Computed , Gemcitabine
13.
Biochem Biophys Res Commun ; 504(2): 393-399, 2018 10 02.
Article in English | MEDLINE | ID: mdl-29890131

ABSTRACT

Arsenic is associated with several adverse health outcomes, and people with diabetes may be more susceptible to arsenic. In this study, we found that arsenic levels in some tissues such as liver, kidney, and heart but not lung of type 1 diabetes mellitus (T1DM) mice were higher than in those of normal mice after a single oral dose of arsenic trioxide for 2 h. However, little is known about the molecular mechanism of the increased tissue uptake of trivalent inorganic arsenic in mice with T1DM. This study aimed to investigate the expression of the mammalian arsenic transporters aquaglyceroporins (AQPs) and glucose transporter 1 (GLUT1) in T1DM mice and compare them with those in normal mice. Results showed that the levels of AQP9 and GLUT1 mRNA and protein were higher in T1DM mouse liver than in the normal one. The levels of AQP7 mRNA and protein were higher in T1DM mouse kidney. In the heart, we observed that the levels of AQP7 and GLUT1 mRNA and protein were higher in T1DM mice, but the levels of AQP9 mRNA and protein in the lung had no significant difference between both mice. These results suggested that T1DM may increase the expression of transporters of trivalent inorganic arsenic and thus increase the arsenic uptake in specific tissues.


Subject(s)
Aquaporins/metabolism , Arsenic/adverse effects , Diabetes Mellitus, Type 1/metabolism , Glucose Transporter Type 1/metabolism , Animals , Arsenic Trioxide/adverse effects , Arsenites/adverse effects , Biological Transport , Blood Glucose/analysis , Body Weight , Inorganic Chemicals , Kidney/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred ICR , RNA, Messenger/metabolism , Tissue Distribution
14.
Chemosphere ; 206: 597-605, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29778937

ABSTRACT

Arsenic (As) is a ubiquitous environmental toxin and robust inducer of oxidative stress (OxS). Copper (Cu) is an essential microelement, which participates in OxS as a cofactor for certain enzymes, with narrow optimal range between essential and toxic concentrations. However, their effects are rarely studied in chicken skeletal muscles, which have soaring per capita consumption andare susceptible to oxidative damage. In the present study, we demonstrated that the administration of copper sulfate (300 mg kg-1) or arsenite (30 mg kg-1) individually or their co-administration leads to varying degrees of OxS in the skeletal muscles of chickens. Corresponding to the protein expression pattern, the mRNA levels of caspase, B-cell lymphoma-2 (Bcl-2) families, and autophagy-related genes were also compromised in the experimental groups, indicating the involvement of both apoptotic and autophagic cell death. Additionally, rampant mitochondrial fission caused the vicious cycle between imbalanced mitochondrial dynamics and OxS, thus tethering intracellular homeostasis. The abovementioned muscle damage and index anomalies were time dependent, and more deteriorated effects were observed in Cu2+ and arsenite co-administered groups than those in groups administered Cu2+ and arsenite alone. Intriguingly, in the studied skeletal muscles, namely wing biceps brachii and leg gastrocnemius, there were conspicuous differences in oxidative toxicity susceptibility, which needs further study. The present study showed that Cu and/or As induce oxidative damage in chicken skeletal muscles and discussed its mechanism in terms of apoptosis, autophagy, and mitochondrial dynamics, thus voicing concerns about poultry breeding areas cross-contaminated with Cu2+ and arsenite.


Subject(s)
Arsenites/adverse effects , Copper/adverse effects , Muscle, Skeletal/metabolism , Oxidative Stress/drug effects , Animals , Apoptosis , Autophagy , Chickens , Mitochondrial Dynamics
15.
Brain Res ; 1690: 23-30, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29630858

ABSTRACT

Optimal cytoplasmic calcium (Ca2+) levels have been associated with adequate cell functioning and neuronal survival. Altered intracellular Ca2+ levels following impaired Ca2+ homeostasis could induce neuronal degeneration or even cell death. There are reports of arsenite induced oxidative stress and the associated disturbances in intracellular calcium homeostasis. The present study focused on determining the strategies that would modulate tissue redox status and calcium binding protein (CaBP) (Calbindin D28k-CB) expression affected adversely by sodium arsenite (NaAsO2) exposure (postnatal) of rat pups. NaAsO2 alone or along with antioxidants (AOXs) (alpha lipoic acid or curcumin) was administered by intraperitoneal (i.p.) route from postnatal day (PND) 1-21 (covering rapid brain growth period - RBGP) to experimental groups and animals receiving sterile water by the same route served as the controls. At the end of the experimental period, the animals were subjected to euthanasia and the cerebellar tissue obtained therefrom was processed for immunohistochemical localization and western blot analysis of CB protein. CB was diffusely expressed in cell body as well as dendritic processes of Purkinje cells (PCs) along the PC Layer (PCL) in all cerebellar folia of the control and the experimental animals. The multilayered pattern of CB +ve cells along with their downregulated expression and low packing density was significantly evident in the arsenic (iAs) alone exposed group as against the controls and AOX supplemented groups. The observations are suggestive of AOX induced restoration of CaBP expression in rat cerebellum following early postnatal exposure to NaAsO2.


Subject(s)
Antioxidants/pharmacology , Arsenites/adverse effects , Calbindins/metabolism , Neuroprotective Agents/pharmacology , Purkinje Cells/drug effects , Sodium Compounds/adverse effects , Animals , Animals, Newborn , Cell Size/drug effects , Curcumin/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Purkinje Cells/metabolism , Purkinje Cells/pathology , Random Allocation , Rats, Wistar , Thioctic Acid/pharmacology , Up-Regulation/drug effects
16.
Biotechnol J ; 13(4): e1700676, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29345424

ABSTRACT

Protein conformational disorders are characterized by disruption of protein folding and toxic accumulation of protein aggregates. Here we describe a sensitive and simple method to follow and monitor general protein aggregation in human cells. Heat shock protein 27 (HSP27) is an oligomeric small heat shock protein that binds and keeps unfolded proteins in a folding competent state. This high specificity of HSP27 for aggregated proteins can be explored to monitor aggregation in living cells by fusing it to a fluorescent protein as Green Fluorescent Protein (GFP). We have constructed a HeLa stable cell line expressing a HSP27:GFP chimeric reporter protein and after validation, this stable cell line is exposed to different agents that interfere with proteostasis, namely Arsenite, MG132, and Aß-peptide. Exposure to proteome destabilizers lead to re-localization of HSP27:GFP fluorescence to foci, confirming that our reporter system is functional and can be used to detect and follow protein aggregation in living cells. This reporter is a valuable tool to setup wide-genetic screens to identify genes and pathways involved in protein misfolding and aggregation.


Subject(s)
Green Fluorescent Proteins/genetics , HSP27 Heat-Shock Proteins/genetics , Proteolysis , Proteome/metabolism , Amyloid beta-Peptides/adverse effects , Arsenites/adverse effects , Green Fluorescent Proteins/metabolism , HSP27 Heat-Shock Proteins/metabolism , HeLa Cells , Heat-Shock Proteins , Humans , Leupeptins/adverse effects , Molecular Chaperones , Protein Aggregates , Protein Binding , Protein Folding , Proteome/chemistry , Recombinant Proteins/metabolism
17.
Biochim Biophys Acta Mol Basis Dis ; 1863(3): 753-763, 2017 03.
Article in English | MEDLINE | ID: mdl-28062277

ABSTRACT

Circular RNAs (circRNAs), a class of noncoding RNAs generated from pre-mRNAs, participate in regulation of genes. The mechanism for regulation, however, is unknown. Here, to determine if, in human keratinocyte (HaCaT) cells, circular RNAs are involved in arsenite-induced acceleration of the cell cycle, a circRNA microarray was performed to analyze the variability of circRNAs in arsenite-treated HaCaT (As-HaCaT) cells and in arsenite-transformed (T-HaCaT) cells in comparison to control HaCaT cells. Among the circRNAs up-regulated in both As-HaCaT cells and T-HaCaT cells, hsa:circRNA_100284 (circ100284) had the greatest increase and was chosen for further research. The presence of circ100284 was confirmed in HaCaT cells. In these cells, arsenite induced increases of EZH2 and cyclin D1 and accelerated the cell cycle. MicroRNA (miR)-217 suppressed the expression of EZH2 was involved in regulation of the cell cycle. Further, in HaCaT cells exposed to arsenite, EZH2 regulated the cell cycle by binding to the promoter of CCND1, which codes for cyclin D1. Moreover, knockdown of circ100284 with siRNA inhibited the cell cycle acceleration induced by arsenite, but this inhibition was reversed by co-transfection with circ100284 siRNA and by a miR-217 inhibitor. Knockdown of circ100284 with siRNA or transfected with miR-217 mimic inhibited the capacity of T-HaCaT cells for colony formation, invasion, and migration, effects that were reversed by co-transfection with a miR-217 inhibitor or by epigenetic expression of EZH2. These results suggest that, in HaCaT cells, arsenite increases circ100284 levels, which act as a sponge for miR-217 and up-regulate the miR-217 target, EZH2, which, in turn, up-regulates cyclin D1and CDK4, and thus accelerates the cell cycle and leads to malignant transformation. Thus, circ100284, via miR-217 regulation of EZH2, is involved in the arsenite-accelerated cell cycle of human keratinocytes in carcinogenesis. This establishes a previously unknown mechanism between arsenite-induced acceleration of the cell cycle and carcinogenesis.


Subject(s)
Arsenites/adverse effects , Carcinogenesis/chemically induced , Carcinogenesis/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Gene Expression Regulation, Neoplastic/drug effects , MicroRNAs/genetics , RNA/genetics , Carcinogenesis/pathology , Cell Cycle/drug effects , Cell Line , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , RNA, Circular
18.
Cancer Lett ; 388: 21-33, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27913196

ABSTRACT

In the cancer microenvironment, extracellular communication allows various types of cells to coordinate and execute biological functions. Emerging evidence indicates that exosomes, as mediators of cell communication, are involved in tumor progression. Little is known, however, about the mechanism by which exosomal miRNAs regulate inflammatory infiltration in arsenite-induced liver cancer. The present research aimed to determine if miRNAs secreted from arsenite-transformed human hepatic epithelial (L-02) cells are transferred into normal L-02 and THLE-3 cells, which are functionally active in the recipient cells. The results show that the exosomes from arsenite-transformed L-02 cells enhance miR-155 expression and the pro-inflammatory properties of normal L-02 and THLE-3 cells. Transformed cells transfer miR-155 into normal L-02 cells via exosomes. The inhibition of NF-κB by siRNA and inhibitor, which reduces miR-155 levels in exosomes derived from transformed L-02 cells, blocks inflammation. Arsenite-transformed cells secrete exosomes to enhance inflammation, but the inhibition of the synthesis of exosomes fails to stimulate inflammation. miR-155 is involved in exosome-mediated intercellular communication between neoplastic and normal liver cells. In addition, miR-155, IL-6, and IL-8 were over-expressed in the serum of arsenite exposure group. And there was a positive correlation between miR-155 and IL-6 or IL-8 levels. Further, exosomal miR-155 was up-regulated in the serum of arsenite exposure group. Thus, these results show that exosomes derived from transformed L-02 cells transfer miR-155 to surrounding cells, which induces pro-inflammatory activity of normal liver cells. The findings support the concept that exosomal miRNAs are involved in cell-cell communication during carcinogenesis induced by environmental chemicals.


Subject(s)
Arsenites/adverse effects , Exosomes/metabolism , Inflammation/chemically induced , MicroRNAs/genetics , NF-kappa B/metabolism , Carcinogenesis/metabolism , Humans , Inflammation/metabolism , Transfection
20.
Wei Sheng Yan Jiu ; 45(1): 93-7, 2016 Jan.
Article in Chinese | MEDLINE | ID: mdl-26987204

ABSTRACT

OBJECTIVE: To investigate the effects of arsenic exposure before and during maternal pregnancy on heart development of fetal rats. METHODS: According to body weight, thirty-two female SD rats (30 to 40 days of age) were randomly divided into control group, low dose group, middle dose group and high dose group with 8 rats per group. They were allowed free access to drinking water with 0, 37.5, 75 and 150 mg/L of sodium arsenite (NaAsO2) for 6 weeks, respectively. Then all the female rats and adult male SD rats were caged together for mating. Once female rats were determined to be pregnant, they would continue to drink deionized distilled water containing different concentrations of sodium arsenite for another 2 weeks. On embryonic day 16, rats were sacrificed to harvest fetuses. Female rats' weight changes, abortions, absorbed fetus number, growth and development of fetal rats were observed. Hematoxylin-eosin staining of serial cardiac slices was performed in embryos to observe cardiac morphology and structure. Fur arsenic contents of female rats were determined with the method of atomic fluorescence spectrometry. RESULTS: Subchronic arsenic exposure caused slow weight growth in female rats. There were two cases of abortion in middle dose group and high dose group, respectively. Compared with these of control group, fetal and placental weight decreased (P < 0.05), and the incidence of fetal absorption increased (P < 0.05) in all arsenic-treated groups. Cardiac malformations in fetal rats including ventricular septal defect, atrial septal defect and tetralogy of Fallot were observed in low, middle and high dose group. The incidence of cardiac malformations increased with the increase of arsenic concentrations in drinking water. Compared with that of control group, the incidence of cardiac malformations remarkably increased in both middle and high dose groups (P < 0.05). Fur arsenic contents increased with the increase of arsenic concentrations in drinking water (P < 0.01). CONCLUSION: Arsenic exposure before and during maternal pregnancy could cause abnormal cardiac development in fetal rats, and increased the risk of congenital heart disease.


Subject(s)
Arsenic/adverse effects , Arsenites/adverse effects , Heart Defects, Congenital/chemically induced , Maternal Exposure/adverse effects , Pregnancy, Animal/metabolism , Sodium Compounds/adverse effects , Animals , Arsenic/analysis , Arsenites/administration & dosage , Body Weight , Drinking Water , Female , Fetal Development , Fetus/metabolism , Male , Placenta/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Reproduction , Sodium Compounds/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...