1
|
Hejazi S, Moosavi M, Molavinia S, Mansouri E, Azadnasab R, Khodayar MJ. Epicatechin ameliorates glucose intolerance and hepatotoxicity in sodium arsenite-treated mice. Food Chem Toxicol 2024; 192:114950. [PMID: 39182636 DOI: 10.1016/j.fct.2024.114950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/04/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Arsenic is a metalloid found in the environment that causes toxic effects in different organs, mainly the liver. This study aimed to investigate the protective effects of epicatechin (EC), a natural flavonol, on glucose intolerance (GI) and liver toxicity caused by sodium arsenite (SA) in mice. Our findings showed that SA exposure led to the development of GI. Liver tissue damage and decreased pancreatic Langerhans islet size were also observed in this study. Mice exposed to SA exhibited hepatic oxidative damage, indicated by reduced antioxidant markers (such as superoxide dismutase, catalase, glutathione peroxidase, and glutathione), along with elevated levels of thiobarbituric acid reactive substances. SA administration elevated the serum activities of liver enzymes alanine aminotransferase, aspartate aminotransferase, and alkaline phosphatase. Furthermore, notable increases in the levels of inflammatory and apoptotic markers (Toll-like receptor 4, nuclear factor-kappa B, tumor necrosis factor-α, nitric oxide, B-cell lymphoma-2, and cysteine aspartate-specific protease-3) were observed in the liver. Treatment of SA-exposed mice with EC considerably reversed these biochemical and histological changes. This study demonstrated the beneficial effects of EC in ameliorating SA-induced hyperglycemia and hepatotoxicity due to its ability to enhance the antioxidant system by modulating inflammation and apoptosis.
Collapse
Affiliation(s)
- Sara Hejazi
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mehrnoosh Moosavi
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shahrzad Molavinia
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Esrafil Mansouri
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Reza Azadnasab
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Javad Khodayar
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
2
|
Rainey NE, Armand AS, Petit PX. Sodium arsenite and arsenic trioxide differently affect the oxidative stress of lymphoblastoid cells: An intricate crosstalk between mitochondria, autophagy and cell death. PLoS One 2024; 19:e0302701. [PMID: 38728286 PMCID: PMC11086853 DOI: 10.1371/journal.pone.0302701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 04/10/2024] [Indexed: 05/12/2024] Open
Abstract
Although the toxicity of arsenic depends on its chemical forms, few studies have taken into account the ambiguous phenomenon that sodium arsenite (NaAsO2) acts as a potent carcinogen while arsenic trioxide (ATO, As2O3) serves as an effective therapeutic agent in lymphoma, suggesting that NaAsO2 and As2O3 may act via paradoxical ways to either promote or inhibit cancer pathogenesis. Here, we compared the cellular response of the two arsenical compounds, NaAsO2 and As2O3, on the Burkitt lymphoma cell model, the Epstein Barr Virus (EBV)-positive P3HR1 cells. Using flow cytometry and biochemistry analyses, we showed that a NaAsO2 treatment induces P3HR1 cell death, combined with drastic drops in ΔΨm, NAD(P)H and ATP levels. In contrast, As2O3-treated cells resist to cell death, with a moderate reduction of ΔΨm, NAD(P)H and ATP. While both compounds block cells in G2/M and affect their protein carbonylation and lipid peroxidation, As2O3 induces a milder increase in superoxide anions and H2O2 than NaAsO2, associated to a milder inhibition of antioxidant defenses. By electron microscopy, RT-qPCR and image cytometry analyses, we showed that As2O3-treated cells display an overall autophagic response, combined with mitophagy and an unfolded protein response, characteristics that were not observed following a NaAsO2 treatment. As previous works showed that As2O3 reactivates EBV in P3HR1 cells, we treated the EBV- Ramos-1 cells and showed that autophagy was not induced in these EBV- cells upon As2O3 treatment suggesting that the boost of autophagy observed in As2O3-treated P3HR1 cells could be due to the presence of EBV in these cells. Overall, our results suggest that As2O3 is an autophagic inducer which action is enhanced when EBV is present in the cells, in contrast to NaAsO2, which induces cell death. That's why As2O3 is combined with other chemicals, as all-trans retinoic acid, to better target cancer cells in therapeutic treatments.
Collapse
Affiliation(s)
- Nathan Earl Rainey
- CNRS UMR 8003 Paris University, SSPIN, Neuroscience Institute, Team “Mitochondria, Apoptosis and Autophagy Signaling”, Campus Saint-Germain, Paris, France
| | - Anne-Sophie Armand
- INSERM U1151, Institut Necker Enfants Malades (INEM), Campus Necker, Université Paris Cité, Paris, France
| | - Patrice X. Petit
- CNRS UMR 8003 Paris University, SSPIN, Neuroscience Institute, Team “Mitochondria, Apoptosis and Autophagy Signaling”, Campus Saint-Germain, Paris, France
| |
Collapse
|
3
|
Vural A, Lanier SM. Properties of biomolecular condensates defined by Activator of G-protein Signaling 3. J Cell Sci 2024; 137:jcs261326. [PMID: 38264908 PMCID: PMC10911133 DOI: 10.1242/jcs.261326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 01/11/2024] [Indexed: 01/25/2024] Open
Abstract
Activator of G-protein signaling 3 (AGS3; also known as GPSM1), a receptor-independent activator of G-protein signaling, oscillates among defined subcellular compartments and biomolecular condensates (BMCs) in a regulated manner that is likely related to the functional diversity of the protein. We determined the influence of cell stress on the cellular distribution of AGS3 and core material properties of AGS3 BMCs. Cellular stress (oxidative, pHi and thermal) induced the formation of AGS3 BMCs in HeLa and COS-7 cells, as determined by fluorescent microscopy. Oxidative stress-induced AGS3 BMCs were distinct from G3BP1 stress granules and from RNA processing BMCs defined by the P-body protein Dcp1a. Immunoblots indicated that cellular stress shifted AGS3, but not the stress granule protein G3BP1 to a membrane pellet fraction following cell lysis. The stress-induced generation of AGS3 BMCs was reduced by co-expression of the signaling protein Gαi3, but not the AGS3-binding partner DVL2. Fluorescent recovery following photobleaching of individual AGS3 BMCs indicated that there are distinct diffusion kinetics and restricted fluidity for AGS3 BMCs. These data suggest that AGS3 BMCs represent a distinct class of stress granules that serve as a previously unrecognized signal processing node.
Collapse
Affiliation(s)
- Ali Vural
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Stephen M. Lanier
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
4
|
Schrenk D, Bignami M, Bodin L, Chipman JK, del Mazo J, Grasl‐Kraupp B, Hogstrand C, Hoogenboom L(R, Leblanc J, Nebbia CS, Nielsen E, Ntzani E, Petersen A, Sand S, Vleminckx C, Wallace H, Barregård L, Benford D, Broberg K, Dogliotti E, Fletcher T, Rylander L, Abrahantes JC, Gómez Ruiz JÁ, Steinkellner H, Tauriainen T, Schwerdtle T. Update of the risk assessment of inorganic arsenic in food. EFSA J 2024; 22:e8488. [PMID: 38239496 PMCID: PMC10794945 DOI: 10.2903/j.efsa.2024.8488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2024] Open
Abstract
The European Commission asked EFSA to update its 2009 risk assessment on arsenic in food carrying out a hazard assessment of inorganic arsenic (iAs) and using the revised exposure assessment issued by EFSA in 2021. Epidemiological studies show that the chronic intake of iAs via diet and/or drinking water is associated with increased risk of several adverse outcomes including cancers of the skin, bladder and lung. The CONTAM Panel used the benchmark dose lower confidence limit based on a benchmark response (BMR) of 5% (relative increase of the background incidence after adjustment for confounders, BMDL05) of 0.06 μg iAs/kg bw per day obtained from a study on skin cancer as a Reference Point (RP). Inorganic As is a genotoxic carcinogen with additional epigenetic effects and the CONTAM Panel applied a margin of exposure (MOE) approach for the risk characterisation. In adults, the MOEs are low (range between 2 and 0.4 for mean consumers and between 0.9 and 0.2 at the 95th percentile exposure, respectively) and as such raise a health concern despite the uncertainties.
Collapse
|
5
|
Jamil M, Mohammadi-Bardbori A, Safa O, Nikpoor AR, Bakhtari A, Mokhtarinejad M, Zadeh SN, Shadboorestan A, Omidi M. Arsenic trioxide-induced cytotoxicity in A549 cells: The role of necroptosis. Drug Res (Stuttg) 2023; 73:417-425. [PMID: 37230480 DOI: 10.1055/a-2076-3246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
INTRODUCTION Lung cancer is one of the deadliest cancers globally. Arsenic trioxide (ATO) is still present as a highly effective drug in treating acute promyelocytic leukemia (APL). Chemotherapy resistance is one of the major problems in cancer therapy. Necroptosis, can overcomes resistance to apoptosis, and can promote cancer treatment. This study examines the necroptosis pathway in A549 cancer cells exposed to ATO. METHODS We used the MTT test to determine the ATO effects on the viability of A549 cells at three different time intervals. Also, the reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) were performed in three-time intervals. The effect of ATO on apoptosis was evaluated by Annexin V / PI staining and, the RIPK1 and MLKL gene expression were measured by Real-Time PCR. RESULTS The ATO has dose and time-dependent cytotoxic effects, so at 24, 48, and 72 h, the IC50 doses were 33.81 '11.44 '2.535 µM respectively. A 50 μM ATO is the most appropriate to increase the MMP loss significantly at all three times. At 24 and 48 h after exposure of cells to ATO, the ROS levels increased. The RIPK1 gene expression increased significantly compared to the control group at concentrations of 50 and 100 μM; however, MLKL gene expression decreased. CONCLUSIONS The A549 cells, after 48 h exposure to ATO at 50 and 100 μM, induces apoptosis and necroptosis. Due to the reduced expression of MLKL, it can be concluded that ATO is probably effective in the metastatic stage of cancer cells.
Collapse
Affiliation(s)
- Maryam Jamil
- Department of Pharmacology & Toxicology, Faculty of Pharmacy and Pharmaceutical Sciences, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Student research committee, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Afshin Mohammadi-Bardbori
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omid Safa
- Department of Clinical Pharmacy, School of Pharmacy and Pharmaceutical Sciences, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Amin Reza Nikpoor
- Depertment of Medical Immunology, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Azizollah Bakhtari
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahnoosh Mokhtarinejad
- Department of Pharmacology & Toxicology, Faculty of Pharmacy and Pharmaceutical Sciences, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Saghar Naybandi Zadeh
- Department of Pharmacognosy and Pharmaceutical Biotechnology, Faculty of Pharmacy and Pharmaceutical Sciences, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Amir Shadboorestan
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahmoud Omidi
- Department of Pharmacology & Toxicology, Faculty of Pharmacy and Pharmaceutical Sciences, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| |
Collapse
|
6
|
Chen ZH, Zhang YL, Yuan B, Chen WJ, Lei GY, Han L. 125I seed implantation enhances arsenic trioxide-induced apoptosis and anti-angiogenesis in lung cancer xenograft mice. Clin Transl Oncol 2023:10.1007/s12094-023-03092-x. [PMID: 36723786 DOI: 10.1007/s12094-023-03092-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 01/17/2023] [Indexed: 02/02/2023]
Abstract
BACKGROUND AND PURPOSE Arsenic trioxide (ATO) exerts anticancer effects on lung cancer. However, the clinical use of ATO is limited due to its systemic toxicity and resistance of lung cancer cells. The present study aimed to investigate the effects of ATO, alone and in combination with 125I seed implantation on tumor growth and proliferation in lung cancer xenograft mice, and investigate the possible molecular mechanisms. METHODS The transmission electron microscope observed the tumor ultrastructure of lung cancer xenograft mice. The proliferation index of Ki-67 and the number and morphology of tumor microvessels were detected with immunohistochemical staining. The protein and mRNA expression were examined by western blot and real-time PCR assay. RESULTS The in vivo results demonstrated that ATO combined with 125I seed significantly inhibited tumor growth and proliferation, as well as promoted apoptosis, and decreased the Ki-67 index and microvessel density in lung cancer xenograft mice. Moreover, ATO combined with 125I seed decreased the protein and mRNA expression levels of HIF-1α, VEGF, and BCL-2, and increased those of BAX and P53. CONCLUSIONS ATO combined with 125I seed significantly inhibited tumor growth and proliferation in lung cancer, which may be accomplished by inhibiting tumor angiogenesis and inducing apoptosis.
Collapse
Affiliation(s)
- Zheng-Hong Chen
- Tumor Hospital of Shaanxi Province, Affiliated to the Medical College of Xi'an Jiaotong University, 309 Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Yi-Li Zhang
- Tumor Hospital of Shaanxi Province, Affiliated to the Medical College of Xi'an Jiaotong University, 309 Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Bin Yuan
- Tumor Hospital of Shaanxi Province, Affiliated to the Medical College of Xi'an Jiaotong University, 309 Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Wen-Juan Chen
- Tumor Hospital of Shaanxi Province, Affiliated to the Medical College of Xi'an Jiaotong University, 309 Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Guang-Yan Lei
- Department of Thoracic Surgery and Seed Therapy Center, Tumor Hospital of Shaanxi Province, Affiliated to the Medical College of Xi'an Jiaotong University, 309 Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China.
| | - Le Han
- Department of Thoracic Surgery and Seed Therapy Center, Tumor Hospital of Shaanxi Province, Affiliated to the Medical College of Xi'an Jiaotong University, 309 Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China.
| |
Collapse
|
7
|
Gandhi D, Bhandari S, Mishra S, Tiwari RR, Rajasekaran S. Non-malignant respiratory illness associated with exposure to arsenic compounds in the environment. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 94:103922. [PMID: 35779705 DOI: 10.1016/j.etap.2022.103922] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 06/22/2022] [Accepted: 06/25/2022] [Indexed: 06/15/2023]
Abstract
Arsenic (As), a toxic metalloid, primarily originates from both natural and anthropogenic activities. Reports suggested that millions of people globally exposed to high levels of naturally occurring As compounds via inhalation and ingestion. There is evidence that As is a well-known lung carcinogen. However, there has been relatively little evidence suggesting its non-malignant lung effects. This review comprehensively summarises current experimental and clinical studies implicating the association of As exposure and the development of several non-malignant lung diseases. Experimental studies provided evidence that As exposure induces redox imbalance, apoptosis, inflammatory response, epithelial-to-mesenchymal transition (EMT), and affected normal lung development through alteration of the components of intracellular signaling cascades. In addition, we also discuss the sources and possible mechanisms of As influx and efflux in the lung. Finally, current experimental studies on treatment strategies using phytochemicals and our perspective on future research with As are also discussed.
Collapse
Affiliation(s)
- Deepa Gandhi
- Department of Biochemistry, ICMR-National Institute for Research in Environmental, Health, Bhopal, Madhya Pradesh, India
| | - Sneha Bhandari
- Department of Biochemistry, ICMR-National Institute for Research in Environmental, Health, Bhopal, Madhya Pradesh, India
| | - Sehal Mishra
- Department of Biochemistry, ICMR-National Institute for Research in Environmental, Health, Bhopal, Madhya Pradesh, India
| | - Rajnarayan R Tiwari
- Department of Biochemistry, ICMR-National Institute for Research in Environmental, Health, Bhopal, Madhya Pradesh, India
| | - Subbiah Rajasekaran
- Department of Biochemistry, ICMR-National Institute for Research in Environmental, Health, Bhopal, Madhya Pradesh, India.
| |
Collapse
|
8
|
Ozturk M, Metin M, Altay V, Bhat RA, Ejaz M, Gul A, Unal BT, Hasanuzzaman M, Nibir L, Nahar K, Bukhari A, Dervash MA, Kawano T. Arsenic and Human Health: Genotoxicity, Epigenomic Effects, and Cancer Signaling. Biol Trace Elem Res 2022; 200:988-1001. [PMID: 33864199 DOI: 10.1007/s12011-021-02719-w] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/11/2021] [Indexed: 02/06/2023]
Abstract
Arsenic is a well-known element because of its toxicity. Humans as well as plants and animals are negatively affected by its exposure. Some countries suffer from high levels of arsenic in their tap water and soils, which is considered a primary arsenic-linked risk factor for living beings. Humans generally get exposed to arsenic by contaminated drinking waters, resulting in many health problems, ranging from cancer to skin diseases. On the other hand, the FDA-certified drug arsenic trioxide provides solutions for various diseases, including several types of cancers. This issue emphasizes the importance of speciation of the metalloid elements in terms of impacts on health. When species get exposed to arsenic, it affects the cells altering their involvement. It can lead to abnormalities in inflammatory mechanisms and the immune system which contribute to the negative impacts generated on the body. The poisoning originating from arsenic gives rise to various biological signs on the body which can be useful for the diagnosis. It is important to find true biomarkers for the detection of arsenic poisoning. In view of its application in medicine and biology, studies on understanding the biological activity of arsenic have increased. In this review, we aim at summarizing the current state of knowledge of arsenic and the mechanism behind its toxicity including genotoxicity, oxidative insults, epigenomic changes, and alterations in cellular signaling.
Collapse
Affiliation(s)
- Munir Ozturk
- Department of Botany and Centre for Environmental Studies, Ege University, Izmir, Turkey.
| | - Mert Metin
- Graduate School of Environmental Engineering, The University of Kitakyushu, 1-1 Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka, 808-0135, Japan
| | - Volkan Altay
- Department of Biology, Faculty of Science and Arts, Hatay Mustafa Kemal University, Hatay, Turkey
| | - Rouf Ahmad Bhat
- Department of Environmental Science, Sri Pratap College, Cluster University Srinagar, Srinagar, Kashmir, India
| | - Mahnoor Ejaz
- Atta-ur-Rahman School of Applied Biosciences, Nat. University of Sciences & Technology, Islamabad, Pakistan
| | - Alvina Gul
- Atta-ur-Rahman School of Applied Biosciences, Nat. University of Sciences & Technology, Islamabad, Pakistan
| | - Bengu Turkyilmaz Unal
- Faculty of Science and Arts, Dept. of Biotechnology, Nigde Omer Halisdemir University, Nigde, Turkey
| | - Mirza Hasanuzzaman
- Department of Agronomy, Faculty of Agriculture, Sher-e-Bangla Agricultural University, Dhaka, Bangladesh
| | - Lutfunnahar Nibir
- Upazilla Health Complex, Ministry of Health, Government of the People's, Homna, Comilla, Bangladesh
| | - Kamuran Nahar
- Dept. of Agricultural Botany, Faculty of Agriculture, Sher-e-Bangla Agricul. University, Dhaka, Bangladesh
| | - Andleep Bukhari
- Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Moonisa Aslam Dervash
- Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, Kashmir, India
| | - Tomonori Kawano
- Graduate School of Environmental Engineering, The University of Kitakyushu, 1-1 Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka, 808-0135, Japan
| |
Collapse
|
9
|
Handali S, Rezaei M. Arsenic and weight loss: At a crossroad between lipogenesis and lipolysis. J Trace Elem Med Biol 2021; 68:126836. [PMID: 34385035 DOI: 10.1016/j.jtemb.2021.126836] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 06/30/2021] [Accepted: 08/02/2021] [Indexed: 12/14/2022]
Abstract
Arsenic is found in soil, food, water and earth crust. Arsenic exposure is associated with chronic diseases such as cancer, cardiovascular disease as well as diabetes. One of complex effects of arsenic is on weight gain or loss. Involvement of arsenic in both weight loss and gain signaling pathways has previously been reported; however, too little attention has been paid to its weight reducing effect. Animal studies exhibited a role of arsenic in weight loss. In this regard, arsenic interference with endocrine system, leptin and adiponectin hormones as well as thermogenesis is more evidence. Apparently, arsenic-induced weight lossis generally meditated by its interaction with thermogenesis. In this review we have discussed the irregularities in metabolic pathways induced by arsenic that can lead to weight loss.
Collapse
Affiliation(s)
- Somayeh Handali
- Medical Biomaterial Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Rezaei
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
10
|
Qu M, Xu H, Li W, Chen J, Zhang Y, Xu B, Li Z, Liu T, Guo L, Xie J. Dynamically monitoring cellular γ-H2AX reveals the potential of carcinogenicity evaluation for genotoxic compounds. Arch Toxicol 2021; 95:3559-3573. [PMID: 34510228 DOI: 10.1007/s00204-021-03156-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 09/02/2021] [Indexed: 02/07/2023]
Abstract
Amongst all toxicological endpoints, carcinogenicity might pose the greatest concern. Genetic damage has been considered an important underlying mechanism for the carcinogenicity of chemical substances. The demand for in vitro genotoxic tests as alternative approaches is growing rapidly with the implementation of new regulations for compounds. However, currently available in vitro genotoxicity tests are often limited by relatively high false positive rates. Moreover, few studies have explored carcinogenicity potential by in vitro genotoxicity testing due to the shortage of suitable toxicological biomarkers to link gene damage with cancer risk. γ-H2AX is a recently acknowledged attractive endpoint (biomarker) for evaluating DNA damage and can simultaneously reflect the DNA damage response and repair of cells. We previously reported an ultrasensitive and reliable method, namely stable-isotope dilution-liquid chromatography-tandem mass spectrometry (ID-LC-MS/MS), for detecting cellular γ-H2AX and evaluating genotoxic chemicals. More importantly, our method can dynamically monitor the specific processes of genotoxic compounds affecting DNA damage and repair reflected by the amount of γ-H2AX. To clarify the possibility of using this method to assess the potential carcinogenicity of genotoxic chemicals, we applied it to a set of 69 model compounds recommended by the European Center for the Validation of Alternative Methods (ECVAM), with already-characterized genotoxic potential. Compared to conventional in vitro genotoxicity assays, including the Ames test, the γ-H2AX assay by MS has high accuracy (94-96%) due to high sensitivity and specificity (88% and 100%, respectively). The dynamic profiles of model compounds after exposure in HepG2 cells were explored, and a mathematical approach was employed to simulate and quantitatively model the DNA repair kinetics of genotoxic carcinogens (GCs) based on γ-H2AX time-effect curves up to 8 h. Two crucial parameters, i.e., k (rate of γ-H2AX decay) and t50 (time required for γ-H2AX from maximum decrease to half) estimated by the least squares method, were achieved. An open web server to help researchers calculate these two key parameters and profile simulated curves of the tested compound is available online ( http://ccb1.bmi.ac.cn:81/shiny-server/sample-apps/prediction1/ ). We detected a positive association between carcinogenic levels and k and t50 values of γ-H2AX in tested GCs, validating the potential of using this MS-based γ-H2AX in vitro assay to help preliminarily evaluate carcinogenicity and assess genotoxicity. This approach may be used alone or integrated into an existing battery of in vitro genetic toxicity tests.
Collapse
Affiliation(s)
- Minmin Qu
- State Key Laboratory of Toxicology and Medical Countermeasures, and Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Hua Xu
- State Key Laboratory of Toxicology and Medical Countermeasures, and Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China.
| | - Wuju Li
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Jia Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, and Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Yajiao Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, and Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Bin Xu
- State Key Laboratory of Toxicology and Medical Countermeasures, and Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Zhi Li
- State Key Laboratory of Toxicology and Medical Countermeasures, and Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Tao Liu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Lei Guo
- State Key Laboratory of Toxicology and Medical Countermeasures, and Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Jianwei Xie
- State Key Laboratory of Toxicology and Medical Countermeasures, and Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China.
| |
Collapse
|
11
|
Ren C, Zhou Y, Liu W, Wang Q. Paradoxical effects of arsenic in the lungs. Environ Health Prev Med 2021; 26:80. [PMID: 34388980 PMCID: PMC8364060 DOI: 10.1186/s12199-021-00998-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 07/15/2021] [Indexed: 11/10/2022] Open
Abstract
High levels (> 100 ug/L) of arsenic are known to cause lung cancer; however, whether low (≤ 10 ug/L) and medium (10 to 100 ug/L) doses of arsenic will cause lung cancer or other lung diseases, and whether arsenic has dose-dependent or threshold effects, remains unknown. Summarizing the results of previous studies, we infer that low- and medium-concentration arsenic cause lung diseases in a dose-dependent manner. Arsenic trioxide (ATO) is recognized as a chemotherapeutic drug for acute promyelocytic leukemia (APL), also having a significant effect on lung cancer. The anti-lung cancer mechanisms of ATO include inhibition of proliferation, promotion of apoptosis, anti-angiogenesis, and inhibition of tumor metastasis. In this review, we summarized the role of arsenic in lung disease from both pathogenic and therapeutic perspectives. Understanding the paradoxical effects of arsenic in the lungs may provide some ideas for further research on the occurrence and treatment of lung diseases.
Collapse
Affiliation(s)
- Caixia Ren
- Department of Respiratory Medicine, The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Yang Zhou
- Liaoning Clinical Research Center for Lung Cancer, The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Wenwen Liu
- Liaoning Clinical Research Center for Lung Cancer, The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Qi Wang
- Department of Respiratory Medicine, The Second Hospital of Dalian Medical University, Dalian, 116023, China.
| |
Collapse
|
12
|
Kuzu M, Kandemir FM, Yıldırım S, Çağlayan C, Küçükler S. Attenuation of sodium arsenite-induced cardiotoxicity and neurotoxicity with the antioxidant, anti-inflammatory, and antiapoptotic effects of hesperidin. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:10818-10831. [PMID: 33099738 DOI: 10.1007/s11356-020-11327-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/18/2020] [Indexed: 06/11/2023]
Abstract
In the scope of the study, the protective effect of hesperidin (HES), a flavanone glycoside, was investigated against sodium arsenite (NaAsO2, SA) induced heart and brain toxicity. For this purpose, 35 Sprague-Dawley male rats were divided into 5 different groups, 7 in each group. Physiological saline was given to the first group. Dose of 200 mg/kg of HES to the second group, 10 mg/kg dose of SA to the 3rd group, 100 mg/kg HES and 10 mg/kg SA to the 4th group, 200 mg/kg HES, and 10 mg/kg SA to the 5th group were given orally for 15 days. At the end of the study, biochemical, histopathological, and immunohistochemical examinations were performed on the heart and brain tissues of the rats. According to the results, SA increased malondialdehyde (MDA) and 8-hydroxy-2'-deoxyguanosine (8-OHdG) levels and decreased glutathione (reduced, GSH) level and superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx) activities in both tissues. Also, it increased cardiac lactate dehydrogenase (LDH) and creatine kinase isoenzyme-MB (CK-MB) activities and cardiac troponin-I level (cTn-I), cerebral acetylcholine esterase activity, nuclear factor kappa-B (NF-κB), tumor necrosis factor-alpha (TNF-α), interleukin-one beta (IL-1β), and cysteine aspartate-specific protease-3 (caspase-3) levels. In addition, as a result of histopathological examination, it was determined that SA damaged tissue architecture, and as a result of immunohistochemical examination, it increased cardiac Bcl-2-associated X protein (Bax) and cerebral glial fibrillary acidic protein (GFAP) expression. The results have also shown that HES co-treatment has an antioxidant, anti-inflammatory, antiapoptotic effect on SA-induced toxicity and aids to protect tissue architecture by showing a regulatory effect on all values. Consequently, it was determined that HES co-treatment had a protective effect on SA-induced heart and brain toxicity in rats.
Collapse
Affiliation(s)
- Müslüm Kuzu
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Karabuk University, Karabuk, Turkey.
| | - Fatih Mehmet Kandemir
- Department of Basic Sciences, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Serkan Yıldırım
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Cüneyt Çağlayan
- Department of Basic Sciences, Faculty of Veterinary Medicine, Bingöl University, Bingol, Turkey
| | - Sefa Küçükler
- Department of Basic Sciences, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| |
Collapse
|
13
|
Das A, Habib G, Vivekanandan P, Kumar A. Reactive oxygen species production and inflammatory effects of ambient PM 2.5 -associated metals on human lung epithelial A549 cells "one year-long study": The Delhi chapter. CHEMOSPHERE 2021; 262:128305. [PMID: 33182158 DOI: 10.1016/j.chemosphere.2020.128305] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 08/29/2020] [Accepted: 09/09/2020] [Indexed: 06/11/2023]
Abstract
The fine particulate matter (PM2.5) was collected at academic campus of Indian Institute of Technology, Delhi, India from January-December 2017. The PM2.5 samples were analysed for carcinogenic (Cd, Cr, As, Ni, and Pb) and non-carcinogenic (V, Cu, Zn, Fe) trace metals and their elicited effects on carcinoma epithelial cell line A549. Toxicological testing was done with ELISA kit. Same analyses were repeated for standard reference material (NIST-1648a) represents urban particulate matter. The student-t test and spearman correlation were used for data analysis. The seasonality in PM2.5 mass concentration and chemical composition showed effect on biological outcomes. The PM2.5 in post-monsoon and winter had higher amount of trace metals compared to mass collected in pre-monsoon and monsoon. Following the trend in PM mass concentration significantly (p < 0.5) lower cell viability was observed in post-monsoon and winter compared to other two seasons. NIST UPM 1648(a) samples always had higher cytotoxicity compared to ambient PM2.5 Delhi sample. Strong association of Chromium, Nickel, Cadmium, and Zinc was observed with cell viability and reactive oxygen species (ROS) production. In winter IL-6, IL-8 production were 2.8 and 3 times higher than values observed in post-monsoon and 53 and 9 times higher than control. In winter season trace metals As, Cu, Fe, in pre-monsoon Cr, Ni, As, Pb, V, and Fe, in post-monsoon Cd and V strongly correlated with ROS generation. ROS production in winter and pre-monsoon seasons found to be 2.6 and 1.3 times higher than extremely polluted post-monsoon season which had 2 to 3 times higher PM2.5 concentration compared to winter and pre-monsoon. The result clearly indicated that the presence of Fe in winter and pre-monsoon seasons catalysed the ROS production, probably OH˙ radical caused high cytokines production which influenced the cell viability reduction, while in post-monsoon PM majorly composed of Pb, As, Fe and Cu and affected by photochemical smog formation showed significant association between ROS production with cell viability. Overall, in Delhi most toxic seasons for respiratory system are winter and post-monsoon and safest season is monsoon.
Collapse
Affiliation(s)
- Ananya Das
- Department of Civil Engineering, Indian Institute of Technology, Delhi, India.
| | - Gazala Habib
- Department of Civil Engineering, Indian Institute of Technology, Delhi, India.
| | - Perumal Vivekanandan
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, India.
| | - Arun Kumar
- Department of Civil Engineering, Indian Institute of Technology, Delhi, India.
| |
Collapse
|
14
|
Islam MM, Takeyama N. Inorganic arsenic administration suppresses human neutrophil function in vitro. Hum Exp Toxicol 2020; 40:725-734. [PMID: 33063558 DOI: 10.1177/0960327120966040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Arsenic, a major environmental toxicant and pollutant, is a global public health concern. Among its many adverse effects, arsenic is immunotoxic, but its effects on human neutrophil functions are not yet well-defined. In this study, we aimed to evaluate the in vitro effects of acute low-dose NaAsO2 exposure on human polymorphonuclear neutrophils (PMNs) for 12 h on the following innate defense mechanisms: formation of neutrophil extracellular traps (NETs), production of reactive oxygen species (ROS), and phagocytosis. Phorbol myristate acetate (PMA) was added to induce NETs formation, which was quantified by measuring cell-free extracellular DNA (cf-DNA), myeloperoxidase-conjugated (MPO)-DNA and neutrophil elastase-conjugated (NE)-DNA, and confirmed by immunofluorescence labeling and imaging. Extracellular bactericidal activity by NETs was evaluated by co-culturing Escherichia coli and PMNs in the presence of a phagocytic inhibitor. Levels of NETs in the culture medium after PMA stimulation was significantly lower in PMNs pre-exposed to arsenic than those not exposed to arsenic. Immunofluorescence staining and extracellular bactericidal activity by NETs revealed similar results. Phagocytosis and ROS production by PMNs were also significantly reduced by arsenic pre-exposure. Together, our findings provide new insights in arsenic immunotoxicity and suggest how it increases susceptibility to infectious diseases in humans.
Collapse
Affiliation(s)
- Md Monirul Islam
- Department of Emergency and Critical Care Medicine, 12703Aichi Medical University, Nagakute, Aichi, Japan
| | - Naoshi Takeyama
- Department of Emergency and Critical Care Medicine, 12703Aichi Medical University, Nagakute, Aichi, Japan
| |
Collapse
|
15
|
Li C, Zhang S, Li L, Hu Q, Ji S. Ursodeoxycholic Acid Protects Against Arsenic Induced Hepatotoxicity by the Nrf2 Signaling Pathway. Front Pharmacol 2020; 11:594496. [PMID: 33178028 PMCID: PMC7596389 DOI: 10.3389/fphar.2020.594496] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
Arsenic is ubiquitous toxic metalloid responsible for many human diseases all over the world. Contrastingly, Ursodeoxycholic acid (UDCA) has been suggested as efficient antioxidant in various liver diseases. However, there are no reports of the effects of UDCA on arsenious acid [As(III)]-induced hepatotoxicity. The objective of this study is to elucidate the protective actions of UDCA on As(III)-induced hepatotoxicity and explore its controlling role in biomolecular mechanisms in vivo and in vitro. The remarkable liver damage induced by As(III) was ameliorated by treatment with UDCA, as reflected by reduced histopathological changes of liver and elevation of serum AST, ALT levels. UDCA play a critical role in stabilization of cellular membrane potential, inhibition of apoptosis and LDH leakage in LO2 cells. Meanwhile, the activities of SOD, CAT and GSH-Px and the level of TSH, GSH were enhanced with UDCA administration, while the accumulations of intracellular ROS, MDA and rate of GSSG/GSH were decreased in vivo and in vitro. Further study disclosed that UDCA significantly inhibited As(III)-induced apoptosis through increasing the expression of Bcl-2 and decreasing the expression of Bax, p53, Cyt C, Cleaved caspase-3 and 9. Moreover, UDCA promoted the expression of nuclear Nrf2, HO-1, and NQO1, although arsenic regulated nuclear translocation of Nrf2 positively. When Nrf2 was silenced, the protective effect of UDCA was abolished. Collectively, the results of this study showed that UDCA protects hepatocytes antagonize As(III)-induced cytotoxicity, and its mechanism may be related to activation of Nrf2 signaling.
Collapse
Affiliation(s)
- Chao Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine, Shanghai Institute for Food and Drug Control, Shanghai, China
| | - Sheng Zhang
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine, Shanghai Institute for Food and Drug Control, Shanghai, China.,School of Pharmacy, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Liming Li
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine, Shanghai Institute for Food and Drug Control, Shanghai, China
| | - Qing Hu
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine, Shanghai Institute for Food and Drug Control, Shanghai, China
| | - Shen Ji
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine, Shanghai Institute for Food and Drug Control, Shanghai, China
| |
Collapse
|
16
|
Wang P, Wan R, Huo W, Dong H, Chang Z, Xia X. Cytotoxicity, genotoxicity, oxidative stress, and apoptosis in HepG2 cells induced by the imidazole ionic liquid 1-dodecyl-3-methylimidazolium chloride. ENVIRONMENTAL TOXICOLOGY 2020; 35:665-672. [PMID: 31916396 DOI: 10.1002/tox.22901] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 12/28/2019] [Accepted: 12/29/2019] [Indexed: 06/10/2023]
Abstract
This study purposes to assess the cytotoxicity of 1-dodecyl-3-methylimidazolium chloride ([C12 min]Cl) in human hepatocellular carcinoma (HepG2) cells. To this end, HepG2 cells were exposed to a range concentration of [C12 min]Cl and evaluated cell viability, genotoxicity, oxidative stress, apoptosis, cell cycle, and apoptosis-related gene expression to determine cytotoxicity. The outcomes showed that [C12 min]Cl curbed HepG2 cell growth and reduced cell viability in a concentration- and time-dependent manner. Moreover, our assay results also revealed that exposure to [C12 min]Cl prompted DNA damage and apoptosis, reduced SOD and GSH content, enhanced MDA level, and changed the cell cycle of HepG2 cells. In addition, [C12 min] Cl caused alters in the expression levels of p53, Bax, and Bcl-2, indicating that p53 and Bcl-2 family may be involved in the cytotoxicity and apoptosis of HepG2 cells induced by [C12 min]C1. In summary, these results indicate that [C12 min]Cl exerts genotoxicity, physiological toxicity and prompts apoptosis in HepG2 cells, and is not an alleged green solvent.
Collapse
Affiliation(s)
- Peijin Wang
- College of Life Science, Henan Normal University, Xinxiang, Henan, People's Republic of China
| | - Ruyan Wan
- College of Life Science, Henan Normal University, Xinxiang, Henan, People's Republic of China
| | - Weiran Huo
- College of Life Science, Henan Normal University, Xinxiang, Henan, People's Republic of China
| | - Hui Dong
- College of Life Science, Henan Normal University, Xinxiang, Henan, People's Republic of China
| | - Zhongjie Chang
- College of Life Science, Henan Normal University, Xinxiang, Henan, People's Republic of China
| | - Xiaohua Xia
- College of Life Science, Henan Normal University, Xinxiang, Henan, People's Republic of China
| |
Collapse
|
17
|
Alpire MES, de Camargo EA, Cardoso CM, Salvadori DMF, Pereira CDS, Ribeiro DA. In vivo and in vitro analysis of cytogenotoxicity in populations living in abnormal conditions from Santos-Sao Vicente estuary. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:12039-12046. [PMID: 31982997 DOI: 10.1007/s11356-020-07602-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 01/01/2020] [Indexed: 06/10/2023]
Abstract
The aim of the study was to evaluate cyto- and genotoxic effects in populations living in subnormal clusters in Santos São Vicente estuary. For in vivo study, samples of buccal mucosa and peripheral blood cells were collected. Micronucleus assay and single-cell gel (comet) assay were performed. For in vitro study, Chinese ovary hamster (CHO) cells were exposed to contaminated water. The results showed that people living in the contaminated estuary have increased DNA damage in oral mucosa and peripheral blood cells, as detected in the micronucleus and comet assays respectively. In addition, estuarine water was able to promote cytotoxicity at the highest concentrations, as well as decrease the number of cells in the G1 phase. In summary, our results indicate that water from the Santos-São Vicente estuary is capable of inducing cytogenotoxicity in mammalian cells in vivo and in vitro.
Collapse
Affiliation(s)
- Maria Esther Suarez Alpire
- Departamento de Biociências, Universidade Federal de São Paulo, UNIFESP, Av. Ana Costa, 95, Vila Mathias, Santos, SP, 11060-001, Brazil
| | | | - Caroline Margonato Cardoso
- Departamento de Biociências, Universidade Federal de São Paulo, UNIFESP, Av. Ana Costa, 95, Vila Mathias, Santos, SP, 11060-001, Brazil
| | | | - Camilo Dias Seabra Pereira
- Department of Sea Sciences, Federal University of São Paulo, UNIFESP, Campus Baixada Santista, Santos, SP, Brazil
| | - Daniel Araki Ribeiro
- Departamento de Biociências, Universidade Federal de São Paulo, UNIFESP, Av. Ana Costa, 95, Vila Mathias, Santos, SP, 11060-001, Brazil.
| |
Collapse
|
18
|
Feng W, Wu X, Mao G, Zhao T, Wang W, Chen Y, Zhang M, Yang L, Wu X. Neurological effects of subchronic exposure to dioctyl phthalate (DOP), lead, and arsenic, individual and mixtures, in immature mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:9247-9260. [PMID: 31916164 DOI: 10.1007/s11356-019-06823-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 10/21/2019] [Indexed: 06/10/2023]
Abstract
Dioctyl phthalate (DOP) (200, 500, and 1000 mg kg-1 bw, i.g.), Pb (Ac)2 (50 mg L-1, p.o.), and NaAsO2 (10 mg L-1, p.o.) were administered individually and as mixtures to weanling male mice for 8 weeks. It was observed that Pb, As, and DOP exposure could significantly inhibit the growth and development of mice. Compared with the Pb, As, and Pb + As groups, the activities of iNOS and TNOS were significantly increased, the levels of AChE and SOD were significantly decreased, and the level of MDA was significantly increased in the Pb + DOP-H, As + DOP-H, and Pb + As + DOP-H groups. The factorial analysis shows that the iNOS, TNOS, and AChE present synergistic effects on Pb, As, and DOP. A significant increase of escape latency and a significant decrease of original platform quadrant stops were observed between Pb + As + DOP-H and Pb + As groups. The factorial analysis shows that there was a synergistic effect on Pb, As, and DOP. Compared with that of the control group, the expression levels of caspase-3 and Bax expression in Pb + As, DOP-H, Pb + DOP-H, As + DOP-H, and Pb + As + DOP-H groups were significantly increased in the hippocampus. The expression levels of Bcl-2 expression decreased significantly and the Bax/Bcl-2 ratio increased significantly. Pathological alterations on the hippocampus were found in exposed groups. This result shows that combined exposure of Pb, As, and DOP could induce neurotoxicity, of which possible mechanism is hippocampal neuronal apoptosis. Graphical abstract This study shows that there were three components with eigenvalues greater than 1, which together explained 89.40% of total variance. The first component (PC1) showed high loadings on B-SOD, L-SOD, B-MDA, L-MDA, K-MDA, iNOS, tNOS, and AChE and accounted for 46.55% of the total variance after Varimax rotation. PC2 accounted for 23.81% of the total variance with high loadings on B-As, L-As, K-As, and K-SOD, whereas PC3 showed high loadings on B-Pb, L-Pb, and K-Pb and accounted for 19.04% of the total variance.
Collapse
Affiliation(s)
- Weiwei Feng
- School of the Environment and Safety Engineering, Jiangsu University, 301 Xuefu Rd., Zhenjiang, 212013, Jiangsu, China
- Institute of Environmental Health and Ecological Security, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xueshan Wu
- School of Chemistry and Chemical Engineering, Jiangsu University, 301 Xuefu Rd., Zhenjiang, 212013, Jiangsu, China
| | - Guanghua Mao
- School of the Environment and Safety Engineering, Jiangsu University, 301 Xuefu Rd., Zhenjiang, 212013, Jiangsu, China
| | - Ting Zhao
- School of Chemistry and Chemical Engineering, Jiangsu University, 301 Xuefu Rd., Zhenjiang, 212013, Jiangsu, China
| | - Wei Wang
- School of Food and Biological Engineering, Jiangsu University, 301 Xuefu Rd., Zhenjiang, 212013, Jiangsu, China
| | - Yao Chen
- School of the Environment and Safety Engineering, Jiangsu University, 301 Xuefu Rd., Zhenjiang, 212013, Jiangsu, China
- Institute of Environmental Health and Ecological Security, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Min Zhang
- School of Chemistry and Chemical Engineering, Jiangsu University, 301 Xuefu Rd., Zhenjiang, 212013, Jiangsu, China
| | - Liuqing Yang
- School of Chemistry and Chemical Engineering, Jiangsu University, 301 Xuefu Rd., Zhenjiang, 212013, Jiangsu, China.
| | - Xiangyang Wu
- School of the Environment and Safety Engineering, Jiangsu University, 301 Xuefu Rd., Zhenjiang, 212013, Jiangsu, China.
- Institute of Environmental Health and Ecological Security, Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
19
|
Cayir A, Barrow TM, Guo L, Byun HM. Exposure to environmental toxicants reduces global N6-methyladenosine RNA methylation and alters expression of RNA methylation modulator genes. ENVIRONMENTAL RESEARCH 2019; 175:228-234. [PMID: 31146095 DOI: 10.1016/j.envres.2019.05.011] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/20/2019] [Accepted: 05/10/2019] [Indexed: 06/09/2023]
Abstract
The epitranscriptome comprises more than 100 forms of RNA modifications. Of these, N6-methyladenosine (m6A) is the most abundantform of RNA methylation, with roles in modulating mRNA transcript processing and regulation. The aims of the study weretoexamine changes inm6A RNA methylation in A549 lung epithelial cells in response to environmental toxicants, anddifferential gene expression of m6A modulator genes ('readers', 'writers' and 'erasers') in human subjects exposed toparticulate matter (PM) and in lung cancer tissueusing publicly-available microarray datasets. Global m6A methylation levelsweremeasured in total RNA after exposuretotwo carcinogens (PM and sodium arsenite) for 24- and 48-h, and totwo endocrine disruptors (bisphenol A and vinclozolin)for 24-h.Global m6A methylation level significantly decreased with exposure to >62 μg/mlPM, >1 μM sodium arsenite, >1 μM bisphenol A (BPA), and0.1 μM vinclozolin. In an analysis of a published dataset derived from a population study, we observed that m6A writers (METTL3 and WTAP), erasers (FTO and ALKBH5) and readers (HNRPC) showed significantly higher expression among participants in the high-PM2.5exposure group compared to those in the low-exposure control group (all p < 0.05). Further, the m6A writer METTL3shows reduced expression in lung tumors in comparison to normal lung epithelia (p < 0.0001). Our findings reveal that m6A RNA methylation can be modified by exposure to environmental toxicants, and exposure to particulate matter is associated with differential expression level of m6A RNA methylation modification machinery.
Collapse
Affiliation(s)
- Akin Cayir
- Vocational Health College, Canakkale Onsekiz Mart University, Canakkale, Turkey; Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, United Kingdom.
| | - Timothy M Barrow
- Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland, United Kingdom
| | - Liqiong Guo
- Tianjin Key Laboratory of Environment, Nutrition and Public Health; Center for International Collaborative Research on Environment, Nutrition and Public Health; Department of Occupational & Environmental Health, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Hyang-Min Byun
- Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, United Kingdom
| |
Collapse
|
20
|
Zhao T, Li X, Sun D, Zhang Z. Oxidative stress: One potential factor for arsenite-induced increase of N 6-methyladenosine in human keratinocytes. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2019; 69:95-103. [PMID: 31004932 DOI: 10.1016/j.etap.2019.04.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 04/03/2019] [Accepted: 04/11/2019] [Indexed: 06/09/2023]
Abstract
N6-methyladenosine (m6A) modification is affected by oxidative stress and gets involved in arsenite toxicity. However, whether oxidative stress is one factor in arsenite-induced alteration of m6A levels remains unclear. Here, reactive oxygen species (ROS), product of lipid peroxidation (MDA), antioxidants (GSH and SOD), m6A levels, m6A methyltransferases (METTL3, METTL14, and WTAP) and demethylases (FTO and ALKBH5) were detected in human keratinocytes exposed to different concentrations of arsenite. Antioxidant N-acetylcysteine was used to assess the influence of arsenite-induced oxidative stress on m6A modification. Possible regulations of m6A modification induced by arsenite were explored using bioinformatic analysis. Our results demonstrated that arsenite-induced oxidative stress increased the levels of m6A methylation possibly by mediating m6A methyltransferases and demethylases, especially elevated expressions of WTAP and METTL14, in human keratinocytes. Whereas N-acetylcysteine suppressed the elevated m6A level and its methyltransferases in human keratinocytes exposed to arsenite. Furthermore, arsenite-induced oxidative stress might mediate m6A methyltransferases and demethylases by reducing transcription of 4 genes (HECTD4, ABCA5, SLC22 A17 and KCNQ5) according to our bioinformatic analysis and experiments. Additionally, GO and Pathway analysis further suggested that the increase of m6A modification in arsenite-induced oxidative stress might be involved in some biological processes such as positive regulation of GTPase activity, apoptotic process, and platelet activation. Taken together, our study revealed the significant role of oxidative stress in m6A modification induced by arsenite.
Collapse
Affiliation(s)
- Tianhe Zhao
- Department of Environmental and Occupational Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| | - Xinyang Li
- Department of Environmental and Occupational Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| | - Donglei Sun
- Department of Environmental and Occupational Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| | - Zunzhen Zhang
- Department of Environmental and Occupational Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
21
|
Zhou J, Ci X, Ma X, Yu Q, Cui Y, Zhen Y, Li S. Pterostilbene Activates the Nrf2-Dependent Antioxidant Response to Ameliorate Arsenic-Induced Intracellular Damage and Apoptosis in Human Keratinocytes. Front Pharmacol 2019; 10:497. [PMID: 31139082 PMCID: PMC6519314 DOI: 10.3389/fphar.2019.00497] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/18/2019] [Indexed: 11/24/2022] Open
Abstract
The NF-E2 p45-related factor 2 (Nrf2), a transcription factor that regulates the cellular adaptive response to oxidative stress, is a target for limiting tissue damage from exposure to environmental toxins, including arsenic. In the current study, we determine whether Pterostilbene (Pts), as a potent activator of Nrf2, has a protective effect on arsenic-induced cytotoxicity and apoptosis in human keratinocytes. Human keratinocytes (HaCaT) or mouse epidermal cells (JB6) were pretreated with Pts for 24 h prior to arsenic treatment. Harvested cells were analyzed by MTT, DCFH-DA, commercial kits, Flow cytometry assay and western blot analysis. Our results demonstrated that Pts effectively regulated the viability in HaCaT and JB6 cells, decreased the reactive oxygen species (ROS) generation and lipid peroxidation (MDA), and improved the NaAsO2-induced depletion of superoxide dismutase (SOD). Moreover, Pts treatment further dramatically inhibited NaAsO2-induced apoptosis, specifically the mitochondrial mediation of apoptosis, which coincided with the effective recovery of NaAsO2-induced mitochondrial membrane potential (ΔΨm) depolarization and cytochrome c release from the mitochondria. Furthermore, arsenic-induced decrease of anti-apoptotic factor Bcl-2 and Bcl-xl, and increase of pro-apoptotic factor Bax and Bad, as well as survival signal related factor caspase 3 activation were reversed by Pts treatment. Further mechanistic studies confirmed that Pts increased antioxidant enzyme expression in a dose-dependent manner, which was related to Nrf2 nuclear translocation. In addition, the effects of Pts on NaAsO2-induced cell viability were largely weakened when Nrf2 was knocked down. Together, our results provide evidence for the use of Pts to activate the Nrf2 pathway to alleviate arsenic-induced dermal damage.
Collapse
Affiliation(s)
- Junfeng Zhou
- Department of Dermatology and Venereology, The First Hospital of Jilin University, Changchun, China
| | - Xinxin Ci
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xiaoyuan Ma
- General Situation of Jilin Provincial Center for Animal Disease Control and Prevention, Jilin University, Changchun, China
| | - Qinlei Yu
- General Situation of Jilin Provincial Center for Animal Disease Control and Prevention, Jilin University, Changchun, China
| | - Yan Cui
- Department of Dermatology and Venereology, The First Hospital of Jilin University, Changchun, China
| | - Yu Zhen
- Department of Dermatology and Venereology, The First Hospital of Jilin University, Changchun, China
| | - Shanshan Li
- Department of Dermatology and Venereology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
22
|
Yu H, Kuang M, Wang Y, Rodeni S, Wei Q, Wang W, Mao D. Sodium Arsenite Injection Induces Ovarian Oxidative Stress and Affects Steroidogenesis in Rats. Biol Trace Elem Res 2019; 189:186-193. [PMID: 30151564 DOI: 10.1007/s12011-018-1467-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/27/2018] [Indexed: 10/28/2022]
Abstract
Oxidative stress is involved in the regulation of mammalian reproduction. The present study was conducted to detect the sodium arsenite-induced oxidative stress and alterations in the structure and steroidogenesis in rat ovary. Twenty female adult rats were injected i.p. with sodium arsenite (8 mg/kg BW, T) or 0.9% saline (C) for 16 days. The oxidative stress indexes and morphology of the liver, kidney, and ovary were detected using commercial kits and HE staining, respectively. The serum progesterone and estradiol were detected by RIA, and the ovarian steroidogenic gene expressions were detected by real-time PCR. Results showed that the ovarian activities of SOD and GSH-PX decreased (P < 0.05), while the ROS activity and MDA level increased (P < 0.05) in the T group. HE staining results showed that treatment with sodium arsenite damaged the ovarian morphology, resulting in reduced large and medium follicles and increased atretic follicles. Nonetheless, neither the liver nor kidney showed evident changes in the oxidative stress indexes or morphology after sodium arsenite treatment. The serum progesterone and estradiol levels decreased (P < 0.05) with the reduced expressions in the ovarian steroidogenic genes (StAR, P450scc, and 3β-HSD) (P < 0.05). In conclusion, sodium arsenite injection can induce ovarian oxidative stress in rats which set up an appropriate model for future studies of ovarian diseases as well as the toxic mechanism of arsenic in the reproduction.
Collapse
Affiliation(s)
- Hao Yu
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Meiqian Kuang
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Yalei Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Saif Rodeni
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Quanwei Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Wei Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Dagan Mao
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, People's Republic of China.
| |
Collapse
|
23
|
Perker MC, Orta Yilmaz B, Yildizbayrak N, Aydin Y, Erkan M. Protective effects of curcumin on biochemical and molecular changes in sodium arsenite‐induced oxidative damage in embryonic fibroblast cells. J Biochem Mol Toxicol 2019; 33:e22320. [DOI: 10.1002/jbt.22320] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/06/2019] [Accepted: 03/13/2019] [Indexed: 11/09/2022]
Affiliation(s)
- Mehmet Can Perker
- Department of BiologyInstitute of Graduate Studies in Sciences, Istanbul UniversityIstanbul Turkey
| | - Banu Orta Yilmaz
- Department of BiologyFaculty of Sciences, Istanbul UniversityIstanbul Turkey
| | - Nebahat Yildizbayrak
- Department of BiologyInstitute of Graduate Studies in Sciences, Istanbul UniversityIstanbul Turkey
| | - Yasemin Aydin
- Department of BiologyFaculty of Sciences, Istanbul UniversityIstanbul Turkey
| | - Melike Erkan
- Department of BiologyFaculty of Sciences, Istanbul UniversityIstanbul Turkey
| |
Collapse
|
24
|
Ma Z, Fan Y, Wu Y, Kebebe D, Zhang B, Lu P, Pi J, Liu Z. Traditional Chinese medicine-combination therapies utilizing nanotechnology-based targeted delivery systems: a new strategy for antitumor treatment. Int J Nanomedicine 2019; 14:2029-2053. [PMID: 30962686 PMCID: PMC6435121 DOI: 10.2147/ijn.s197889] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cancer is a major public health problem, and is now the world’s leading cause of death. Traditional Chinese medicine (TCM)-combination therapy is a new treatment approach and a vital therapeutic strategy for cancer, as it exhibits promising antitumor potential. Nano-targeted drug-delivery systems have remarkable advantages and allow the development of TCM-combination therapies by systematically controlling drug release and delivering drugs to solid tumors. In this review, the anticancer activity of TCM compounds is introduced. The combined use of TCM for antitumor treatment is analyzed and summarized. These combination therapies, using a single nanocarrier system, namely codelivery, are analyzed, issues that require attention are determined, and future perspectives are identified. We carried out a systematic review of >280 studies published in PubMed since 1985 (no patents involved), in order to provide a few basic considerations in terms of the design principles and management of targeted nanotechnology-based TCM-combination therapies.
Collapse
Affiliation(s)
- Zhe Ma
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ; .,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ;
| | - Yuqi Fan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ; .,School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yumei Wu
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ; .,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ;
| | - Dereje Kebebe
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ; .,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ; .,School of Pharmacy, Institute of Health Sciences, Jimma University, Jimma, Ethiopia
| | - Bing Zhang
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ; .,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ;
| | - Peng Lu
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ; .,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ;
| | - Jiaxin Pi
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ; .,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ;
| | - Zhidong Liu
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ; .,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ;
| |
Collapse
|
25
|
Rezaei M, Keshtzar E, Khodayar MJ, Javadipour M. SirT3 regulates diabetogenic effects caused by arsenic: An implication for mitochondrial complex II modification. Toxicol Lett 2019; 301:24-33. [DOI: 10.1016/j.toxlet.2018.10.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/27/2018] [Accepted: 10/22/2018] [Indexed: 01/11/2023]
|
26
|
Naz S, Battu S, Khan RA, Afroz S, Giddaluru J, Vishwakarma SK, Satti V, Habeeb MA, Khan AA, Khan N. Activation of integrated stress response pathway regulates IL-1β production through posttranscriptional and translational reprogramming in macrophages. Eur J Immunol 2019; 49:277-289. [PMID: 30578631 DOI: 10.1002/eji.201847513] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 10/27/2018] [Accepted: 12/10/2018] [Indexed: 01/06/2023]
Abstract
Immune cells sense and programme its cellular machinery appropriately to the environmental changes through the activation of cytoprotective adaptive pathway so-called the "integrated stress response (ISR)". However, the mechanisms implicated in ISR-induced protective responses are poorly understood. Here, we show that ISR activation by arsenite (Ar) results in suppression of IL-1β production in macrophages and inhibition of DSS-induced colitis in a murine model through a novel posttranscriptional and translation regulatory (PTR) mechanism. Ar triggers PTR events through eIF2α-phosphorylation, which results in the attenuation of active polysome formation leading to the accumulation of translationally stalled IL-1β mRNAs. Translationally stalled IL-1β mRNAs recruit RNA-binding proteins (TIA-1/TIAR), resulting in the formation of RBP-RNA complexes known as stress granules (SGs). The SGs bound IL-1β mRNAs might undergo degradation through induction of autophagy. Also, we show that Ar posttranslationally impairs processing and secretion of IL-1β by diminishing inflammasome activation. Altogether, this study unveils a novel mechanism of IL-1β regulation and further suggests that pharmacological activation of cytoprotective ISR pathway might provide an effective therapeutic intervention against inflammatory diseases.
Collapse
Affiliation(s)
- Saima Naz
- Central Laboratory for Stem Cell Research and Translational Medicine, Center for Liver Research and Diagnostics, Deccan College of Medical Sciences, Hyderabad, India
| | - Srikanth Battu
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Rafiq Ahmad Khan
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Sumbul Afroz
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Jeevan Giddaluru
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Sandeep Kumar Vishwakarma
- Central Laboratory for Stem Cell Research and Translational Medicine, Center for Liver Research and Diagnostics, Deccan College of Medical Sciences, Hyderabad, India
| | | | - Md Aejaz Habeeb
- Central Laboratory for Stem Cell Research and Translational Medicine, Center for Liver Research and Diagnostics, Deccan College of Medical Sciences, Hyderabad, India
| | - Aleem Ahmed Khan
- Central Laboratory for Stem Cell Research and Translational Medicine, Center for Liver Research and Diagnostics, Deccan College of Medical Sciences, Hyderabad, India
| | - Nooruddin Khan
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, India
| |
Collapse
|
27
|
Roy JS, Chatterjee D, Das N, Giri AK. Substantial Evidences Indicate That Inorganic Arsenic Is a Genotoxic Carcinogen: a Review. Toxicol Res 2018; 34:311-324. [PMID: 30370006 PMCID: PMC6195883 DOI: 10.5487/tr.2018.34.4.311] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 08/24/2018] [Accepted: 09/07/2018] [Indexed: 01/04/2023] Open
Abstract
Arsenic is one of the most toxic environmental toxicants. More than 150 million people worldwide are exposed to arsenic through ground water contamination. It is an exclusive human carcinogen. Although the hallmarks of arsenic toxicity are skin lesions and skin cancers, arsenic can also induce cancers in the lung, liver, kidney, urinary bladder, and other internal organs. Arsenic is a non-mutagenic compound but can induce significant cytogenetic damage as measured by chromosomal aberrations, sister chromatid exchanges, and micronuclei formation in human systems. These genotoxic end points are extensively used to predict genotoxic potentials of different environmental chemicals, drugs, pesticides, and insecticides. These cytogenetic end points are also used for evaluating cancer risk. Here, by critically reviewing and analyzing the existing literature, we conclude that inorganic arsenic is a genotoxic carcinogen.
Collapse
Affiliation(s)
- Jinia Sinha Roy
- Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Debmita Chatterjee
- Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Nandana Das
- Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Ashok K Giri
- Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
28
|
Li X, Li L, Huang Y, Liu B, Chi H, Shi L, Zhang W, Li G, Niu Y, Zhu X. Synergistic therapy of chemotherapeutic drugs and MTH1 inhibitors using a pH-sensitive polymeric delivery system for oral squamous cell carcinoma. Biomater Sci 2018; 5:2068-2078. [PMID: 28744533 DOI: 10.1039/c7bm00395a] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
MutT homolog 1 (MTH1) is an essential sanitizer of the free nucleotide pool that prevents lethal DNA damage in cancer cells, which has been validated as an anticancer target in recent years. Small molecule TH287 potently and selectively inhibits the MTH1 protein in cells. Here, we developed an effective chemotherapeutic system for oral squamous cell carcinoma (OSCC) based on polymeric nanoparticles that achieve co-delivery of anticancer drug sodium arsenite (NaAsO2) and MTH1 inhibitor TH287. Cationic hyperbranched poly(amine-ester) (HPAE), an amphiphilic and pH-sensitive polymer with a highly branched structure, self-assembled into nanoparticles in aqueous solution. Both NaAsO2 and TH287 could be loaded into HPAE nanoparticles with the help of electrostatic attraction and hydrophobic interaction. The release of NaAsO2 and TH287 from HPAE(NaAsO2 + TH287) nanoparticles was pH-dependent. In vitro evaluation demonstrated that the HPAE(NaAsO2 + TH287) nanoparticles rapidly entered cancer cells and released NaAsO2 and TH287 in response to acidic intracellular environments. In comparison with NaAsO2, TH287, HPAE(NaAsO2) nanoparticles, HPAE(TH287) nanoparticles, and the physical mixture of HPAE(NaAsO2) nanoparticles and TH287, the HPAE(NaAsO2 + TH287) nanoparticles exhibited more effective inhibition of tumor cell proliferation, illustrating the synergistic effect of NaAsO2 and TH287. The experimental results show that TH287 is likely to inhibit MTH1 in tumor cells, rendering them more sensitive to NaAsO2.
Collapse
Affiliation(s)
- Xiao Li
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Lisek K, Campaner E, Ciani Y, Walerych D, Del Sal G. Mutant p53 tunes the NRF2-dependent antioxidant response to support survival of cancer cells. Oncotarget 2018; 9:20508-20523. [PMID: 29755668 PMCID: PMC5945496 DOI: 10.18632/oncotarget.24974] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 03/09/2018] [Indexed: 12/15/2022] Open
Abstract
NRF2 (NFE2L2) is one of the main regulators of the antioxidant response of the cell. Here we show that in cancer cells NRF2 targets are selectively upregulated or repressed through a mutant p53-dependent mechanism. Mechanistically, mutant p53 interacts with NRF2, increases its nuclear presence and resides with NRF2 on selected ARE containing gene promoters activating the transcription of a specific set of genes while leading to the transcriptional repression of others. We show that thioredoxin (TXN) is a mutant p53-activated NRF2 target with pro-survival and pro-migratory functions in breast cancer cells under oxidative stress, while heme oxygenase 1 (HMOX1) is a mutant p53-repressed target displaying opposite effects. A gene signature of NRF2 targets activated by mutant p53 shows a significant association with bad overall prognosis and with mutant p53 status in breast cancer patients. Concomitant inhibition of thioredoxin system with Auranofin and of mutant p53 with APR-246 synergizes in killing cancer cells expressing p53 gain-of-function mutants.
Collapse
Affiliation(s)
- Kamil Lisek
- National Laboratory CIB, Area Science Park Padriciano, Trieste 34149, Italy.,Present address: Max-Delbrück-Centrum for Molecular Medicine, Berlin 13092, Germany
| | - Elena Campaner
- National Laboratory CIB, Area Science Park Padriciano, Trieste 34149, Italy.,Department of Life Sciences, University of Trieste, Trieste 34127, Italy
| | - Yari Ciani
- National Laboratory CIB, Area Science Park Padriciano, Trieste 34149, Italy
| | - Dawid Walerych
- National Laboratory CIB, Area Science Park Padriciano, Trieste 34149, Italy.,Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Giannino Del Sal
- National Laboratory CIB, Area Science Park Padriciano, Trieste 34149, Italy.,Department of Life Sciences, University of Trieste, Trieste 34127, Italy
| |
Collapse
|
30
|
Elia AC, Magara G, Caruso C, Masoero L, Prearo M, Arsieni P, Caldaroni B, Dörr AJM, Scoparo M, Salvati S, Brizio P, Squadrone S, Abete MC. A comparative study on subacute toxicity of arsenic trioxide and dimethylarsinic acid on antioxidant status in Crandell Rees feline kidney (CRFK), human hepatocellular carcinoma (PLC/PRF/5), and epithelioma papulosum cyprini (EPC) cell lines. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2018; 81:333-348. [PMID: 29498595 DOI: 10.1080/15287394.2018.1442758] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 02/15/2018] [Accepted: 02/15/2018] [Indexed: 05/29/2023]
Abstract
Arsenic (As) is a global contaminant of terrestrial and aquatic environments posing concern for environmental and human health. The effects of subacute concentrations of arsenic trioxide (AsIII) and dimethylarsinic acid (DMAV) were examined using Crandell Rees feline kidney (CRFK), human hepatocellular carcinoma (PLC/PRF/5), and epithelioma papulosum cyprini (EPC). Whole monolayer with suffering cells (confluence 100%, pyknosis and refractive cells; value scale = 2) led to identification of subacute As concentrations for the three cell lines. The selected AsIII concentrations were 1.33 µM for CRFK and 33.37 µM for PLC/PRF/5 and EPC, at 48 hr time point. The selected DMAV concentrations were 0.67 mM for PLC/PRF/5, 1.33 mM for CRFK, and 2.67 mM for EPC for 48 hr. Unlike the AsIII test, the three cell lines did not exhibit marked susceptibility to DMAV-mediated toxicity. Several oxidative stress biomarker levels, directly or indirectly associated with reactive oxygen species (ROS) elimination including superoxide dismutase, catalase, glutathione peroxidases, glutathione reductase, glutathione S-transferase, glyoxalase I, glyoxalase II, and total glutathione, were determined in the three cell lines at 24 and 48 hr. Antioxidant responses in metal-treated cells were significantly altered compared to controls, suggesting a perturbation of redox state. The weakening of antioxidant pathway in either healthy or tumoral cells was greater using AsIII than DMAV. Differences in level of several oxidative stress biomarkers suggest that the oxidative stress mechanism induced by AsIII is distinctly different from DMAV. Multifaceted mechanisms of action underlying ROS generation in tumor and nontumor cells versus AsIII and DMAV exposure are thus involved. Since As-mediated toxicity is quite complex, more data regarding both oxidant-enhancement and oxidant-lowering strategies may be useful to improve knowledge regarding the influence of As on human and animal cells.
Collapse
Affiliation(s)
- Antonia Concetta Elia
- a Dipartimento di Chimica, Biologia e Biotecnologie , Università degli Studi di Perugia , Perugia , Italy
| | - Gabriele Magara
- a Dipartimento di Chimica, Biologia e Biotecnologie , Università degli Studi di Perugia , Perugia , Italy
| | - Claudio Caruso
- b Istituto Zooprofilattico Sperimentale del Piemonte , Torino , Italy
| | - Loretta Masoero
- b Istituto Zooprofilattico Sperimentale del Piemonte , Torino , Italy
| | - Marino Prearo
- b Istituto Zooprofilattico Sperimentale del Piemonte , Torino , Italy
| | - Paola Arsieni
- b Istituto Zooprofilattico Sperimentale del Piemonte , Torino , Italy
| | - Barbara Caldaroni
- a Dipartimento di Chimica, Biologia e Biotecnologie , Università degli Studi di Perugia , Perugia , Italy
| | | | - Melissa Scoparo
- a Dipartimento di Chimica, Biologia e Biotecnologie , Università degli Studi di Perugia , Perugia , Italy
| | - Stefania Salvati
- b Istituto Zooprofilattico Sperimentale del Piemonte , Torino , Italy
| | - Paola Brizio
- b Istituto Zooprofilattico Sperimentale del Piemonte , Torino , Italy
| | | | | |
Collapse
|
31
|
Minatel BC, Sage AP, Anderson C, Hubaux R, Marshall EA, Lam WL, Martinez VD. Environmental arsenic exposure: From genetic susceptibility to pathogenesis. ENVIRONMENT INTERNATIONAL 2018; 112:183-197. [PMID: 29275244 DOI: 10.1016/j.envint.2017.12.017] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/15/2017] [Accepted: 12/12/2017] [Indexed: 05/21/2023]
Abstract
More than 200 million people in 70 countries are exposed to arsenic through drinking water. Chronic exposure to this metalloid has been associated with the onset of many diseases, including cancer. Epidemiological evidence supports its carcinogenic potential, however, detailed molecular mechanisms remain to be elucidated. Despite the global magnitude of this problem, not all individuals face the same risk. Susceptibility to the toxic effects of arsenic is influenced by alterations in genes involved in arsenic metabolism, as well as biological factors, such as age, gender and nutrition. Moreover, chronic arsenic exposure results in several genotoxic and epigenetic alterations tightly associated with the arsenic biotransformation process, resulting in an increased cancer risk. In this review, we: 1) review the roles of inter-individual DNA-level variations influencing the susceptibility to arsenic-induced carcinogenesis; 2) discuss the contribution of arsenic biotransformation to cancer initiation; 3) provide insights into emerging research areas and the challenges in the field; and 4) compile a resource of publicly available arsenic-related DNA-level variations, transcriptome and methylation data. Understanding the molecular mechanisms of arsenic exposure and its subsequent health effects will support efforts to reduce the worldwide health burden and encourage the development of strategies for managing arsenic-related diseases in the era of personalized medicine.
Collapse
Affiliation(s)
- Brenda C Minatel
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Adam P Sage
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Christine Anderson
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Roland Hubaux
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Erin A Marshall
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Wan L Lam
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Victor D Martinez
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada.
| |
Collapse
|
32
|
Dodson M, de la Vega MR, Harder B, Castro-Portuguez R, Rodrigues SD, Wong PK, Chapman E, Zhang DD. Low-level arsenic causes proteotoxic stress and not oxidative stress. Toxicol Appl Pharmacol 2018; 341:106-113. [PMID: 29408041 PMCID: PMC5929483 DOI: 10.1016/j.taap.2018.01.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 01/12/2018] [Accepted: 01/21/2018] [Indexed: 01/22/2023]
Abstract
Prolonged exposure to arsenic has been shown to increase the risk of developing a number of diseases, including cancer and type II diabetes. Arsenic is present throughout the environment in its inorganic forms, and the level of exposure varies greatly by geographical location. The current recommended maximum level of arsenic exposure by the EPA is 10μg/L, but levels>50-1000μg/L have been detected in some parts of Asia, the Middle East, and the Southwestern United States. One of the most important steps in developing treatment options for arsenic-linked pathologies is to understand the cellular pathways affected by low levels of arsenic. Here, we show that acute exposure to non-lethal, low-level arsenite, an environmentally relevant arsenical, inhibits the autophagy pathway. Furthermore, arsenite-induced autophagy inhibition initiates a transient, but moderate ER stress response. Significantly, low-level arsenite exposure does not exhibit an increase in oxidative stress. These findings indicate that compromised autophagy, and not enhanced oxidative stress occurs early during arsenite exposure, and that restoring the autophagy pathway and proper proteostasis could be a viable option for treating arsenic-linked diseases. As such, our study challenges the existing paradigm that oxidative stress is the main underlying cause of pathologies associated with environmental arsenic exposure.
Collapse
Affiliation(s)
- Matthew Dodson
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721
| | - Montserrat Rojo de la Vega
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721
| | - Bryan Harder
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721
| | - Raul Castro-Portuguez
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721
| | - Silvia D. Rodrigues
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721
| | - Pak Kin Wong
- Department of Biomedical Engineering, The Pennsylvania State University, University, Park, PA, USA, 16802
| | - Eli Chapman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721
| | - Donna D. Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA, 85721,Arizona Cancer Center, University of Arizona, Tucson, AZ, USA, 85724,To whom correspondence should be addressed: Dr. Donna D. Zhang, College of Pharmacy, 1703 East Mabel Street, Tucson, AZ, 85721, , Phone number: 520-626-9918
| |
Collapse
|
33
|
Jiang X, Chen C, Gu S, Zhang Z. Regulation of ABCG2 by nuclear factor kappa B affects the sensitivity of human lung adenocarcinoma A549 cells to arsenic trioxide. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 57:141-150. [PMID: 29274627 DOI: 10.1016/j.etap.2017.12.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 11/18/2017] [Accepted: 12/16/2017] [Indexed: 06/07/2023]
Abstract
Arsenic trioxide (As2O3) is successfully used as an anticancer agent against acute promyelocytic leukemia and some solid tumors. However, the application of As2O3 is largely limited by its drug resistance in the treatment of non-small cell lung carcinoma (NSCLC). Therefore, it is an urgent task to enhance the sensitivity of lung cancer cells to As2O3. In this study, using human lung adenocarcinoma A549 cells as a cell culture model, we demonstrated that an adenosine triphosphate binding cassette (ABC) transporter, ABCG2, was significantly increased by As2O3 treatment, while other ABC transporters, ABCB1 and ABCC1 showed no remarkable change in the response to As2O3. After inhibition of ABCG2 by its specific inhibitor, the drug sensitivity of As2O3 to A549 cells was significantly enhanced, manifested by decreased cell viability and colony formation as well as the increased ROS production and cell apoptosis. To further understand the molecular mechanism underlying the elevation of ABCG2 expression in As2O3-treated cells, we detected the activation state of nuclear factor kappa B (NF-κB) pathway and its relationship with ABCG2 expression. Our results revealed that the increased expression of ABCG2 was regulated by NF-κB, and thus affecting the cell death of As2O3-treated A549 cells. These findings indicate that inhibition of NF-κB/ABCG2 pathway by specific inhibitors may be a new strategy for the improvement of As2O3 sensitivity in NSCLC treatment.
Collapse
Affiliation(s)
- Xuejun Jiang
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China; Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, People's Republic of China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China; Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing, People's Republic of China
| | - Shiyan Gu
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Zunzhen Zhang
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
34
|
Chen H, Gu S, Dai H, Li X, Zhang Z. Dihydroartemisinin Sensitizes Human Lung Adenocarcinoma A549 Cells to Arsenic Trioxide via Apoptosis. Biol Trace Elem Res 2017; 179:203-212. [PMID: 28261759 DOI: 10.1007/s12011-017-0975-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 02/15/2017] [Indexed: 11/29/2022]
Abstract
Recent studies have shown that arsenic trioxide (ATO) is an effective anti-cancer drug for treatment of acute promyelocytic leukemia and other types of human cancer. However, we have found that lung cancer cells constantly develop a high level of resistance to ATO. In this study, we have explored a possibility of combination of dihydroartemisinin (DHA) and ATO treatments to reduce ATO resistance of lung cancer cells. We determined the combinatory effects of DHA and ATO on cytotoxicity of human lung adenocarcinoma (A549) cells. We showed that co-exposure to DHA and ATO of A549 cells synergistically increased the cytotoxicity and apoptotic cell death in the cells. We found that the synergistic effect of DHA and ATO in promoting apoptosis mainly resulted from increased cellular level of reactive oxygen species (ROS) and DNA damage. ATO alone only exerted moderate growth inhibitory effects on A549 cells. The results indicate that DHA can significantly sensitize ATO-induced cytotoxicity of A549 lung cancer cells through apoptosis mediated by ROS-induced DNA damage. Interestingly, we found that the combinatory treatment of DHA and ATO did not result in significant adverse effects in normal human bronchial epithelial (HBE) cells. Our results further provide evidence for the potential application of combinatory effects of DHA and ATO as a safe therapy for human lung cancer.
Collapse
Affiliation(s)
- Hongyu Chen
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Shiyan Gu
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Huangmei Dai
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xinyang Li
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Zunzhen Zhang
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
35
|
Gu S, Lai Y, Chen H, Liu Y, Zhang Z. miR-155 mediates arsenic trioxide resistance by activating Nrf2 and suppressing apoptosis in lung cancer cells. Sci Rep 2017; 7:12155. [PMID: 28939896 PMCID: PMC5610328 DOI: 10.1038/s41598-017-06061-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/06/2017] [Indexed: 12/16/2022] Open
Abstract
Arsenic trioxide (ATO) resistance is a challenging problem in chemotherapy. However, the underlying mechanisms remain to be elucidated. In this study, we identified a high level of expression of miR-155 in a human lung adenocarcinoma A549R cell line that is highly resistant to ATO. We showed that the high level of miR-155 was associated with increased levels of cell survival, colony formation, cell migration and decreased cellular apoptosis, and this was mediated by high levels of Nrf2, NAD(P)H quinone oxidoreductase 1 (NQO1), heme oxygenase-1 (HO-1) and a high ratio of Bcl-2/Bax. Overexpression of the miR-155 mimic in A549R cells resulted in increased levels of colony formation and cell migration as well as reduced apoptosis along with increased Nrf2, NQO1 and HO-1. In contrast, silencing of miR-155 expression with its inhibitor in the cells, significantly decreased the cellular levels of Nrf2, NQO1 and HO-1 as well as the ratio of Bcl-2/Bax. This subsequently reduced the level of colony formation and cell migration facilitating ATO-induced apoptosis. Our results indicate that miR-155 mediated ATO resistance by upregulating the Nrf2 signaling pathway, but downregulating cellular apoptosis in lung cancer cells. Our study provides new insights into miR-155-mediated ATO resistance in lung cancer cells.
Collapse
Affiliation(s)
- Shiyan Gu
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yanhao Lai
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, 33199, USA
| | - Hongyu Chen
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yuan Liu
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, 33199, USA. .,Biochemistry Ph.D. Program, Florida International University, Miami, Florida, 33199, USA. .,Biomolecular Sciences Institute, Florida International University, Miami, Florida, 33199, USA.
| | - Zunzhen Zhang
- Department of Environmental Health and Occupational Medicine, West China School of Public Health, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
36
|
Guidarelli A, Fiorani M, Cerioni L, Scotti M, Cantoni O. Arsenite induces DNA damage via mitochondrial ROS and induction of mitochondrial permeability transition. Biofactors 2017; 43:673-684. [PMID: 28703385 DOI: 10.1002/biof.1375] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 06/16/2017] [Accepted: 06/21/2017] [Indexed: 02/06/2023]
Abstract
Arsenite is an established DNA-damaging agent and human carcinogen. We initially selected conditions in which the metalloid causes DNA strand scission in the absence of detectable apoptotic DNA degradation in U937 cells. This response was suppressed by catalase and by treatments (rotenone and ascorbic acid), or manipulations (respiration-deficient phenotype), preventing the mitochondrial formation of O2-. ( mitoO2-.). MitoO2-., and its dismutation product, H2 O2 , are therefore critically involved in the arsenite-dependent DNA-damaging response. We then established a link between mitoO2-./H2 O2 and mitochondrial permeability transition (MPT), and found that this second event also promoted the formation of DNA-damaging species. As a consequence, the DNA damage induced by arsenite, in addition to being abolished by the aforementioned treatments/manipulations, was also significantly reduced by the MPT inhibitor cyclosporin A (CsA). A CsA-sensitive induction of p53 mRNA expression was also detected. Finally, evidence of CsA-sensitive DNA strand scission was also obtained in MCF-7, HT22, and NCTC-2544 cells. MitoO2-./H2 O2 therefore directly mediates DNA damage induced by arsenite and indirectly promotes the formation of additional DNA-damaging species via the induction of MPT. © 2017 BioFactors, 43(5):673-684, 2017.
Collapse
Affiliation(s)
- Andrea Guidarelli
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", 61029, Urbino, Italy
| | - Mara Fiorani
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", 61029, Urbino, Italy
| | - Liana Cerioni
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", 61029, Urbino, Italy
| | - Maddalena Scotti
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", 61029, Urbino, Italy
| | - Orazio Cantoni
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", 61029, Urbino, Italy
| |
Collapse
|
37
|
Chen QY, Costa M. A comprehensive review of metal-induced cellular transformation studies. Toxicol Appl Pharmacol 2017; 331:33-40. [DOI: 10.1016/j.taap.2017.05.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/30/2017] [Accepted: 05/05/2017] [Indexed: 01/07/2023]
|
38
|
Chen C, Jiang X, Gu S, Lai Y, Liu Y, Zhang Z. Protection of Nrf2 against arsenite-induced oxidative damage is regulated by the cyclic guanosine monophosphate-protein kinase G signaling pathway. ENVIRONMENTAL TOXICOLOGY 2017; 32:2004-2020. [PMID: 27774770 PMCID: PMC5403658 DOI: 10.1002/tox.22374] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 10/03/2016] [Accepted: 10/05/2016] [Indexed: 05/05/2023]
Abstract
Arsenite has been shown to induce a variety of oxidative damage in mammalian cells. However, the mechanisms underlying cellular responses to its adverse effects remain unknown. We previously showed that the level of Nrf2, a nuclear transcription factor significantly increased in arsenite-treated human bronchial epithelial (HBE) cells suggesting that Nrf2 is involved in responding to arsenite-induced oxidative damage. To explore how Nrf2 can impact arsenite-induced oxidative damage, in this study, we examined Nrf2 activation and its regulation upon cellular arsenite exposure as well as its effects on arsenite-induced oxidative damage in HBE cells. We found that Nrf2 mRNA and protein levels were significantly increased by arsenite in a dose- and time-dependent manner. Furthermore, we showed that over-expression of Nrf2 significantly reduced the level of arsenite-induced oxidative damage in HBE cells including DNA damage, chromosomal breakage, lipid peroxidation and depletion of antioxidants. This indicates a protective role of Nrf2 against arsenite toxicity. This was further supported by the fact that activation of Nrf2 by its agonists, tertiary butylhydroquinone (t-BHQ) and sulforaphane (SFN) resulted in the same protective effects against arsenite toxicity. Moreover, we demonstrated that arsenite-induced activation of Nrf2 was mediated by the cyclic guanosine monophosphate (cGMP)-protein kinase G (PKG) signaling pathway. This is the first evidence showing that Nrf2 protects against arsenite-induced oxidative damage through the cGMP-PKG pathway. Our study suggests that activation of Nrf2 through the cGMP-PKG signaling pathway in HBE cells may be developed as a new strategy for prevention of arsenite toxicity. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 2004-2020, 2017.
Collapse
Affiliation(s)
- Chengzhi Chen
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People’s Republic of China
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing, People’s Republic of China
| | - Xuejun Jiang
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People’s Republic of China
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing, People’s Republic of China
| | - Shiyan Gu
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Yanhao Lai
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, USA
| | - Yuan Liu
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, USA
- Biochemistry Ph.D. Program, Florida International University, Miami, Florida, USA
- Biomolecular Sciences Institute, Florida International University, Miami, Florida, USA
- Corresponding authors: Zunzhen Zhang, Ph.D., Department of Environmental Health, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu 610041, People’s Republic of China. ; Tel: +86 028 85501298; Fax: +86 028 85501295, Yuan Liu, Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8 Street, Miami, FL, 33199, USA ; Tel: 305-348-3628; Fax: 305-348-3772
| | - Zunzhen Zhang
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People’s Republic of China
- Corresponding authors: Zunzhen Zhang, Ph.D., Department of Environmental Health, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu 610041, People’s Republic of China. ; Tel: +86 028 85501298; Fax: +86 028 85501295, Yuan Liu, Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8 Street, Miami, FL, 33199, USA ; Tel: 305-348-3628; Fax: 305-348-3772
| |
Collapse
|
39
|
Chen C, Jiang X, Gu S, Zhang Z. MicroRNA-155 regulates arsenite-induced malignant transformation by targeting Nrf2-mediated oxidative damage in human bronchial epithelial cells. Toxicol Lett 2017; 278:38-47. [PMID: 28688901 DOI: 10.1016/j.toxlet.2017.07.215] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/01/2017] [Accepted: 07/04/2017] [Indexed: 12/23/2022]
Abstract
Arsenite is a well-documented human lung carcinogen but the detailed mechanisms of carcinogenesis remain unclear. In this study, human bronchial epithelial (16-HBE) cells were continuously exposed to 2.5μM arsenite for about 13 weeks to induce the phenotypes of malignant transformation. Our results showed that Nrf2 expression was gradually decreased whereas no significant change was observed on NF-κB activation with increased time of arsenite exposure. To test the roles of Nrf2-meidtaed oxidative damage in the arsenite-induced malignant transformation, we compared the levels of cGMP, PKG and oxidative damage-related indicators between arsenic-transformed cells and control cells. Our data demonstrated there were no significantly differences on the contents of cGMP, PKG, MDA and the production of ROS, but the levels of GSH and NO, the activities of SOD, tNOS and iNOS were significantly enhanced in the arsenic-transformed cells. Importantly, Nrf2 inactivation could be modulated by miR-155, and inhibition of miR-155 remarkably attenuated the malignant phenotypes and promoted apoptotic cell death in the arsenic-transformed cells. Together, our findings provide the novel mechanism that miR-155 may regulate arsenite-induced cell malignant transformation by targeting Nrf2-mediated oxidative damage, indicating that inhibition of miR-155 may be a potential strategy against lung carcinogenesis of arsenite.
Collapse
Affiliation(s)
- Chengzhi Chen
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China; Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing, People's Republic of China
| | - Xuejun Jiang
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China; Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, People's Republic of China
| | - Shiyan Gu
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Zunzhen Zhang
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
40
|
Oyagbemi AA, Omobowale TO, Asenuga ER, Ochigbo GO, Adejumobi AO, Adedapo AA, Yakubu MA. Sodium arsenite-induced cardiovascular and renal dysfunction in rat via oxidative stress and protein kinase B (Akt/PKB) signaling pathway. Redox Rep 2017; 22:467-477. [DOI: 10.1080/13510002.2017.1308910] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Ademola Adetokunbo Oyagbemi
- Department of Veterinary Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | | | | | - Grace Onyeche Ochigbo
- Department of Veterinary Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | | | - Adeolu Alex Adedapo
- Department of Veterinary Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Momoh Audu Yakubu
- Department of Environmental and Interdisciplinary Sciences, COSET, Texas Southern University, Houston, TX, USA
| |
Collapse
|
41
|
Molecular insight of arsenic-induced carcinogenesis and its prevention. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:443-455. [PMID: 28229170 DOI: 10.1007/s00210-017-1351-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 01/26/2017] [Indexed: 12/20/2022]
Abstract
Population of India and Bangladesh and many other parts of the world are badly exposed to arsenic through drinking water. Due to non-availability of safe drinking water, they are dependent on arsenic-contaminated water. Generally, poverty level is high in those areas with lack of proper nutrition. Arsenic is considered to be an environmental contaminant and widely distributed in the environment due to its natural existence and anthropogenic applications. Contamination of arsenic in both human and animal could occur through air, soil, and other sources. Arsenic exposure mainly occurs in food materials through drinking water with high levels of arsenic in it. High levels of arsenic in groundwater have been found to be associated with various health-related problems including arsenicosis, skin lesions, cardiovascular diseases, reproductive problems, psychological, neurological, immunotoxic, and carcinogenesis. The mechanism of arsenic toxicity consists in its transformation in metaarsenite, which acylates protein sulfhydryl groups, affect on mitochondria by inhibiting succinic dehydrogenase activity and can uncouple oxidative phosphorylation with production of active oxygen species by tissues. A variety of dietary antioxidant supplements are useful to protect the carcinogenetic effects of arsenic. They play crucial role for counteracting oxidative damage and protect carcinogenesis by chelating with heavy metal moiety. Phytochemicals and chelating agents will be beneficial for combating heavy metal-induced carcinogenesis through its biopharmaceutical properties.
Collapse
|
42
|
Adegoke AM, Gbadegesin MA, Odunola OA. Methanol Extract of Adansonia digitata Leaf Protects Against Sodium Arsenite-induced Toxicities in Male Wistar Rats. Pharmacognosy Res 2017; 9:7-11. [PMID: 28250647 PMCID: PMC5330107 DOI: 10.4103/0974-8490.199768] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Human and animal population exposure to arsenic through the consumption of arsenic contaminated water is rampant in many parts of the world. Protective agents of medicinal plants origin could provide maximum protection against toxicities of various kinds. OBJECTIVE The protective role of orally administered methanol extract of the leaves of Adansonia digitata (MELAD) on sodium arsenite (SA) - induced clastogenicity and hepatotoxicity in male Wistar rats was evaluated. MATERIALS AND METHODS Thirty male Wistar rats divided into six Groups (1-6) of five animals each were used for the study. Group 1 (negative control) received distilled water and normal diet only, Groups 2-6 received the extract (at 250 or 500 mg/kg body weight) and/or SA at 2.5 mg/kg body weight. RESULTS There was statistically significant (P < 0.05) increase in the number of micronucleated polychromatic erythrocytes and lipid peroxidation in the SA group as compared with the negative control and treated groups. Administration of the extract reduced the effects of SA on the above parameters. Activities of serum alanine and aspartate aminotransferases did not show statistically significant effects; however, the histological analyses revealed periportal cellular infiltration by mononuclear cells, whereas the MELAD treated groups show mild cellular infiltration and mild portal congestion. CONCLUSIONS MELAD protect against SA-induced toxicities in rats, and it may offer protection in circumstances of co-exposure and cases of arsenicosis. SUMMARY MELAD extract significantly reduce the lipid peroxidation induced by sodium arsenite in the liver of rats.MELAD did not show profound effects on the activities of serum alanine (ALT) and aspartate (AST) aminotranferases.MELAD offered significant protection against sodium arsenite-induced genotoxicity in the micronuclei induction assay.In the circumstances of co-exposure to arsenic contamination, MELAD may protect against sodium arsenite-induced toxicities. Abbreviations Used: MELAD: Methanol extract of the leaves of Adansonia digitata, SA: Sodium arsenite, nMPCEs: Number of micronucleated polychromatic erythocytes; ALT: Alanine aminotranferase; AST: Aspartate aminotranferase, TBARS: Thiobarbituric acid reactive substances, TBA: Thiobarbituric acid, MDA: malondialdehyde, Sodium arsenite (NaAsO2), IARC: International Agency for Research on Cancer.
Collapse
Affiliation(s)
- Ayodeji Mathias Adegoke
- Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Michael Adedapo Gbadegesin
- Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Oyeronke Adunni Odunola
- Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
43
|
Howe CG, Gamble MV. Influence of Arsenic on Global Levels of Histone Posttranslational Modifications: a Review of the Literature and Challenges in the Field. Curr Environ Health Rep 2016; 3:225-37. [PMID: 27352015 PMCID: PMC4967376 DOI: 10.1007/s40572-016-0104-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Arsenic is a human carcinogen and also increases the risk for non-cancer outcomes. Arsenic-induced epigenetic dysregulation may contribute to arsenic toxicity. Although there are several reviews on arsenic and epigenetics, these have largely focused on DNA methylation. Here, we review investigations of the effects of arsenic on global levels of histone posttranslational modifications (PTMs). Multiple studies have observed that arsenic induces higher levels of H3 lysine 9 dimethylation (H3K9me2) and also higher levels of H3 serine 10 phosphorylation (H3S10ph), which regulate chromosome segregation. In contrast, arsenic causes a global loss of H4K16ac, a histone PTM that is a hallmark of human cancers. Although the findings for other histone PTMs have not been entirely consistent across studies, we discuss biological factors which may contribute to these inconsistencies, including differences in the dose, duration, and type of arsenic species examined; the tissue or cell line evaluated; differences by sex; and exposure timing. We also discuss two important considerations for the measurement of histone PTMs: proteolytic cleavage of histones and arsenic-induced alterations in histone expression.
Collapse
Affiliation(s)
- Caitlin G. Howe
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University. Address: 11 Floor, 722 W. 168 Street, New York, New York, 10032. . Phone: 212-305-1205. Fax: 212-305-3857
| | - Mary V. Gamble
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University. Address: 11 Floor, 722 W. 168 Street, New York, New York, 10032. . Phone: 212-305-7949. Fax: 212-305-3857
| |
Collapse
|
44
|
Rahman S, Housein Z, Dabrowska A, Mayán MD, Boobis AR, Hajji N. E2F1-mediated FOS induction in arsenic trioxide-induced cellular transformation: effects of global H3K9 hypoacetylation and promoter-specific hyperacetylation in vitro. ENVIRONMENTAL HEALTH PERSPECTIVES 2015; 123:484-92. [PMID: 25574600 PMCID: PMC4421767 DOI: 10.1289/ehp.1408302] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 01/06/2015] [Indexed: 05/21/2023]
Abstract
BACKGROUND Aberrant histone acetylation has been observed in carcinogenesis and cellular transformation associated with arsenic exposure; however, the molecular mechanisms and cellular outcomes of such changes are poorly understood. OBJECTIVE We investigated the impact of tolerated and toxic arsenic trioxide (As2O3) exposure in human embryonic kidney (HEK293T) and urothelial (UROtsa) cells to characterize the alterations in histone acetylation and gene expression as well as the implications for cellular transformation. METHODS Tolerated and toxic exposures of As2O3 were identified by measurement of cell death, mitochondrial function, cellular proliferation, and anchorage-independent growth. Histone extraction, the MNase sensitivity assay, and immunoblotting were used to assess global histone acetylation levels, and gene promoter-specific interactions were measured by chromatin immunoprecipitation followed by reverse-transcriptase polymerase chain reaction. RESULTS Tolerated and toxic dosages, respectively, were defined as 0.5 μM and 2.5 μM As2O3 in HEK293T cells and 1 μM and 5 μM As2O3 in UROtsa cells. Global hypoacetylation of H3K9 at 72 hr was observed in UROtsa cells following tolerated and toxic exposure. In both cell lines, tolerated exposure alone led to H3K9 hyperacetylation and E2F1 binding at the FOS promoter, which remained elevated after 72 hr, contrary to global H3K9 hypoacetylation. Thus, promoter-specific H3K9 acetylation is a better predictor of cellular transformation than are global histone acetylation patterns. Tolerated exposure resulted in an increased expression of the proto-oncogenes FOS and JUN in both cell lines at 72 hr. CONCLUSION Global H3K9 hypoacetylation and promoter-specific hyperacetylation facilitate E2F1-mediated FOS induction in As2O3-induced cellular transformation.
Collapse
Affiliation(s)
- Sunniyat Rahman
- Centre for Pharmacology and Therapeutics, Department of Medicine, Imperial College London, London, United Kingdom
| | | | | | | | | | | |
Collapse
|
45
|
Chen C, Jiang X, Lai Y, Liu Y, Zhang Z. Resveratrol protects against arsenic trioxide-induced oxidative damage through maintenance of glutathione homeostasis and inhibition of apoptotic progression. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2015; 56:333-46. [PMID: 25339131 PMCID: PMC4376608 DOI: 10.1002/em.21919] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 10/07/2014] [Indexed: 05/22/2023]
Abstract
Arsenic trioxide (As2 O3 ) is commonly used to treat acute promyelocytic leukemia and solid tumors. However, the clinical application of the agent is limited by its cyto- and genotoxic effects on normal cells. Thus, relief of As2 O3 toxicity in normal cells is essentially necessary for improvement of As2 O3 -mediated chemotherapy. In this study, we have identified a series of protective effects of resveratrol against As2 O3 -induced oxidative damage in normal human bronchial epithelial (HBE) cells. We showed that treatment of HBE cells with resveratrol significantly reduced cellular levels of DNA damage, chromosomal breakage, and apoptosis induced by As2 O3 . The effect of resveratrol against DNA damage was associated with a decreased level of reactive oxygen species and lipid peroxidation in cells treated by As2 O3 , suggesting that resveratrol protects against As2 O3 toxicity via a cellular anti-oxidative stress pathway. Further analysis of the roles of resveratrol demonstrated that it modulated biosynthesis, recycling, and consumption of glutathione (GSH), thereby promoting GSH homeostasis in HBE cells treated by As2 O3 . This was further supported by results showing that resveratrol prevented an increase in the activities and levels of caspases, Fas, Fas-L, and cytochrome c proteins induced by As2 O3 . Our study indicates that resveratrol relieves As2 O3 -induced oxidative damage in normal human lung cells via maintenance of GSH homeostasis and suppression of apoptosis.
Collapse
Affiliation(s)
- Chengzhi Chen
- Department of Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Xuejun Jiang
- Department of Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yanhao Lai
- Department of Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, USA
| | - Yuan Liu
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, USA
- Biomolecular Sciences Institute, Florida International University, Miami, Florida, USA
- Corresponding authors: Zunzhen Zhang, M.D., Ph.D., Department of Environmental Health, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu 610041, People's Republic of China. ; ; Tel: +86 028 85501298; Fax: +86 028 85501295, Yuan Liu, M.D., Ph.D., Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8 Street, Miami, Florida, 33199, USA. ; Tel: 305-348-3628
| | - Zunzhen Zhang
- Department of Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China
- Corresponding authors: Zunzhen Zhang, M.D., Ph.D., Department of Environmental Health, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu 610041, People's Republic of China. ; ; Tel: +86 028 85501298; Fax: +86 028 85501295, Yuan Liu, M.D., Ph.D., Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8 Street, Miami, Florida, 33199, USA. ; Tel: 305-348-3628
| |
Collapse
|
46
|
Gu S, Chen C, Jiang X, Zhang Z. Resveratrol synergistically triggers apoptotic cell death with arsenic trioxide via oxidative stress in human lung adenocarcinoma A549 cells. Biol Trace Elem Res 2015; 163:112-23. [PMID: 25431299 DOI: 10.1007/s12011-014-0186-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 11/17/2014] [Indexed: 10/24/2022]
Abstract
Arsenic trioxide (As2O3) is a potent anticancer drug for the treatment of acute promyelocytic leukemia. However, the clinical applications of the agent to treat solid tumors are largely compromised by the drug resistance. Our previous study demonstrated that resveratrol, a plant-derived natural product, could potentiate the toxicity of arsenite in lung adenocarcinoma A549 cells at relatively high concentration, indicating that combination of resveratrol and As2O3 may be a helpful strategy to solve the drug resistance of As2O3 in tumor cells. To test this possibility, in the present study, we determined the combined effects of resveratrol and As2O3 in cultured A549 cells. Our results showed that co-treatment of resveratrol with As2O3 resulted in a synergistic augmentation of cytotoxicity and apoptosis in cells at the tested concentration. To further reveal the detailed mechanism of this synergistic effect on cytotoxicity and apoptosis, apoptosis-related proteins, DNA and chromosomal damage, and the level of oxidative stress were also evaluated. Our data revealed that co-treatment with resveratrol and As2O3 caused more genotoxicity and serious oxidative stress in A549 cells than that of single agent treatment. Moreover, resveratrol and As2O3 could also corporately enhance the release of cytochrome c and the expressions of death receptor Fas and FasL. Together, our results suggest that resveratrol and As2O3 synergistically increase the apoptotic cell death in A549 cells through induction of oxidative stress, indicating that the combination of resveratrol with As2O3 may be a promising strategy to increase the clinical efficacy of As2O3 in the treatment of lung tumor.
Collapse
Affiliation(s)
- Shiyan Gu
- Department of Environmental Health, West China School of Public Health, Sichuan University, No. 16, Section 3, Renmin Nan Road, Chengdu, 610041, People's Republic of China
| | | | | | | |
Collapse
|
47
|
Luo Q, Li Y, Deng J, Zhang Z. PARP-1 inhibitor sensitizes arsenic trioxide in hepatocellular carcinoma cells via abrogation of G2/M checkpoint and suppression of DNA damage repair. Chem Biol Interact 2014; 226:12-22. [PMID: 25499136 DOI: 10.1016/j.cbi.2014.12.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Revised: 11/16/2014] [Accepted: 12/03/2014] [Indexed: 02/06/2023]
Abstract
Arsenic trioxide (ATO) is successfully used to treat hematological malignancies. However, the clinical application of the agent in solid tumors is largely limited by its dose-dependent toxicity which results from the high intrinsic resistance of the cancer cells. In this study, we firstly identified a series of sensitization effects of 4AN, a PARP-1 inhibitor, on human hepatocellular carcinoma cell line HepG2 to ATO treatment. We showed that treatment of HepG2 cells with 4AN promoted ATO-induced cell death in a synergistic manner. The ATO-sensitization by 4AN was associated with its effect on abrogation of ATO-induced G2/M checkpoint which impairs DNA damage repair and promotes cell apoptosis. Further analysis demonstrated that the ATO-induced G2/M checkpoint was closely related to a decrease in cyclin B1, a key G2/M mediator; whereas 4AN up-regulated the expression of cyclin B1 in ATO-treated cells, which may be at least partly responsible for its effect on abrogation of ATO-induced G2/M checkpoint. This was further supported by the result showing that down-regulation of cyclin B1 using siRNA could restore the G2/M checkpoint in cells co-treated with ATO and 4AN, thereby improving DNA damage repair and decreasing apoptosis. Our study indicates that the abrogation of G2/M checkpoint and the suppression of DNA damage repair contribute to ATO-sensitization by PARP-1 inhibitor in HepG2 cells, which provides a novel insight into the chemo-sensitization mechanism of PARP-1 inhibitor.
Collapse
Affiliation(s)
- Qingying Luo
- Department of Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yang Li
- Department of Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jianjun Deng
- Department of Laboratory, Mianyang 404 Hospital, Mianyang, Sichuan, People's Republic of China
| | - Zunzhen Zhang
- Department of Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
48
|
Mohanta RK, Garg AK, Dass RS, Behera SK. Blood biochemistry, thyroid hormones, and oxidant/antioxidant status of guinea pigs challenged with sodium arsenite or arsenic trioxide. Biol Trace Elem Res 2014; 160:238-44. [PMID: 24948398 DOI: 10.1007/s12011-014-0041-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 06/09/2014] [Indexed: 10/25/2022]
Abstract
The present experiment aimed to compare the two most commonly used compounds of arsenic (sodium arsenite and arsenic trioxide) for their effect on blood metabolites, thyroid hormones, and oxidant/antioxidant status in guinea pigs. Twenty-one adult guinea pigs were randomly divided into three equal groups. Animals in group T1 (control) were fed a basal diet, whereas 50 ppm arsenic was added in the basal diet either as sodium arsenite (T2) or arsenic trioxide (T3) and fed for 11 weeks. Serum aspartate aminotransferase and alanine aminotransferase activities were significantly increased along with a decrease in blood hemoglobin level in both the arsenic-administered groups. The level of erythrocytic antioxidants (catalase, superoxide dismutase, reduced glutathione, glutathione-S-transferase, and glutathione reductase) was decreased and lipid peroxidation was elevated upon arsenic exposure. Serum thyroid hormone levels were reduced and arsenic levels in tissues increased in both the arsenic-exposed groups, irrespective of the arsenic compound. Thus, sodium arsenite and arsenic trioxide exerted similar adverse effects on blood metabolic profile, antioxidant status, and thyroid hormones in guinea pigs.
Collapse
Affiliation(s)
- Ranjan Kumar Mohanta
- Animal Nutrition Division, Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, 243 122, India,
| | | | | | | |
Collapse
|
49
|
Critical role of cellular glutathione homeostasis for trivalent inorganic arsenite-induced oxidative damage in human bronchial epithelial cells. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2014; 770:35-45. [DOI: 10.1016/j.mrgentox.2014.04.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 04/28/2014] [Accepted: 04/29/2014] [Indexed: 12/19/2022]
|
50
|
Srivastava P, Yadav RS, Chandravanshi LP, Shukla RK, Dhuriya YK, Chauhan LKS, Dwivedi HN, Pant AB, Khanna VK. Unraveling the mechanism of neuroprotection of curcumin in arsenic induced cholinergic dysfunctions in rats. Toxicol Appl Pharmacol 2014; 279:428-440. [PMID: 24952339 DOI: 10.1016/j.taap.2014.06.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 06/06/2014] [Accepted: 06/07/2014] [Indexed: 12/20/2022]
Abstract
Earlier, we found that arsenic induced cholinergic deficits in rat brain could be protected by curcumin. In continuation to this, the present study is focused to unravel the molecular mechanisms associated with the protective efficacy of curcumin in arsenic induced cholinergic deficits. Exposure to arsenic (20mg/kg body weight, p.o) for 28 days in rats resulted to decrease the expression of CHRM2 receptor gene associated with mitochondrial dysfunctions as evident by decrease in the mitochondrial membrane potential, activity of mitochondrial complexes and enhanced apoptosis both in the frontal cortex and hippocampus in comparison to controls. The ultrastructural images of arsenic exposed rats, assessed by transmission electron microscope, exhibited loss of myelin sheath and distorted cristae in the mitochondria both in the frontal cortex and hippocampus as compared to controls. Simultaneous treatment with arsenic (20mg/kg body weight, p.o) and curcumin (100mg/kg body weight, p.o) for 28 days in rats was found to protect arsenic induced changes in the mitochondrial membrane potential and activity of mitochondrial complexes both in frontal cortex and hippocampus. Alterations in the expression of pro- and anti-apoptotic proteins and ultrastructural damage in the frontal cortex and hippocampus following arsenic exposure were also protected in rats simultaneously treated with arsenic and curcumin. The data of the present study reveal that curcumin could protect arsenic induced cholinergic deficits by modulating the expression of pro- and anti-apoptotic proteins in the brain. More interestingly, arsenic induced functional and ultrastructural changes in the brain mitochondria were also protected by curcumin.
Collapse
Affiliation(s)
- Pranay Srivastava
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Rajesh S Yadav
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India; Department of Crimnology and Forensic Science, Harisingh Gour University, Sagar 470 003, India
| | - Lalit P Chandravanshi
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Rajendra K Shukla
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Yogesh K Dhuriya
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Lalit K S Chauhan
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Hari N Dwivedi
- Babu Banarasi Das University, BBD City, Faizabad Road, Lucknow 227 015, India
| | - Aditiya B Pant
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India
| | - Vinay K Khanna
- CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow 226 001, India.
| |
Collapse
|