1
|
Manuguerra S, Carli F, Scoditti E, Santulli A, Gastaldelli A, Messina CM. Effects of Mixtures of Emerging Pollutants and Drugs on Modulation of Biomarkers Related to Toxicity, Oxidative Stress, and Cancer. Metabolites 2024; 14:559. [PMID: 39452940 PMCID: PMC11509268 DOI: 10.3390/metabo14100559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/03/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Background/Objectives: Over time, the scientific community has developed a growing interest in the effects of mixtures of different compounds, for which there is currently no established evidence or knowledge, in relation to certain categories of xenobiotics. It is well known that exposure to pollutants causes oxidative stress, resulting in the overproduction of reactive oxygen species (ROS), which can affect signaling pathways that regulate the cell cycle, apoptosis, energy balance, and cellular metabolism. The aim of this study was to investigate the effects of sub-lethal concentrations of mixtures of emerging pollutants and pharmaceuticals on the modulation of biomarkers related to toxicity, oxidative stress, and cancer. Methods: In this study, the hepatoma cell line HepG2 was exposed to increasing concentrations of polybrominated diphenyl ether 47 (BDE-47), cadmium chloride (CdCl2), and carbamazepine (CBZ), both individually and in mixtures, for 72 h to assess cytotoxicity using the MTT assay. The subsequent step, following the identification of the sub-lethal concentration, was to investigate the effects of exposure at the gene expression level, through the evaluation of molecular markers related to cell cycle and apoptosis (p53), oxidative stress (NRF2), conjugation and detoxification of xenobiotics (CYP2C9 and GST), DNA damage (RAD51 and γH2AFX), and SUMOylation processes (SUMO1 and UBC9) in order to identify any potential alterations in pathways that are normally activated at the cellular level. Results: The results showed that contaminants tend to affect the enzymatic detoxification and antioxidant system, influencing DNA repair defense mechanisms involved in resistance to oxidative stress. The combined effect of the compounds at sub-lethal doses results in a greater activation of these pathways compared to exposure to each compound alone, thereby exacerbating their cytotoxicity. Conclusions: The biomarkers analyzed could contribute to the definition of early warning markers useful for environmental monitoring, while simultaneously providing insight into the toxicity and hazard levels of these substances in the environment and associated health risks.
Collapse
Affiliation(s)
- Simona Manuguerra
- Laboratory of Marine Biochemistry and Ecotoxicology, Department of Earth and Marine Sciences DiSTeM, University of Palermo, Via G. Barlotta 4, 91100 Trapani, Italy; (S.M.); (A.S.)
| | - Fabrizia Carli
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy;
| | - Egeria Scoditti
- Institute of Clinical Physiology, National Research Council, 73100 Lecce, Italy;
| | - Andrea Santulli
- Laboratory of Marine Biochemistry and Ecotoxicology, Department of Earth and Marine Sciences DiSTeM, University of Palermo, Via G. Barlotta 4, 91100 Trapani, Italy; (S.M.); (A.S.)
| | - Amalia Gastaldelli
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy;
| | - Concetta Maria Messina
- Laboratory of Marine Biochemistry and Ecotoxicology, Department of Earth and Marine Sciences DiSTeM, University of Palermo, Via G. Barlotta 4, 91100 Trapani, Italy; (S.M.); (A.S.)
| |
Collapse
|
2
|
Yang Y, Gao C, Li Q, Liu Y, Cao J. HMGA2-mediated glutamine metabolism is required for Cd-induced cell growth and cell migration. Toxicology 2024; 507:153899. [PMID: 39032683 DOI: 10.1016/j.tox.2024.153899] [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: 03/31/2024] [Revised: 07/01/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Cadmium (Cd) exposure significantly increases the risk of lung cancer. The demand for glutamine is increasing in cancers, including lung cancer. In this study, we investigated the role of glutamine metabolism in Cd-induced cell growth and migration. Firstly, we found that 2 μM Cd-treatment up-regulated the expression of ASCT2 (alanine, serine, cysteine-preferring transporter 2) and ASNS (asparagine synthetase) while downregulating mitochondrial glutaminase GLS1 in A549 cells. The same results were obtained in male BALB/c mice treated with 0.5 and 1 mg Cd/kg body weight. Subsequently, both glutamine deprivation and transfection with siASCT2 revealed that glutamine played a role in Cd-induced cell growth and migration. Furthermore, using 4-PBA (5 mM), an inhibitor of endoplasmic reticulum (ER) stress, Tm (0.1 μg/ml), an inducer of ER stress, siHMGA2, and over-expressing HMGA2 plasmids we demonstrated that ER stress/HMGA2 axis was involved in inducing ASCT2 and ASNS, while inhibiting GLS1. Additionally, the chromatin immunoprecipitation assay using an HMGA2 antibody revealed the direct binding of the HMGA2 to the promoter sequences of the ASCT2, ASNS, and GLS1 genes. Finally, dual luciferase reporter assay determined that HMGA2 increased the transcription of ASCT2 and ASNS while inhibiting the transcription of GLS1. Overall, we found that ER stress-induced HMGA2 controls glutamine metabolism by transcriptional regulation of ASCT2, ASNS and GLS1 to accelerate cell growth and migration during exposure to Cd at low concentrations. This study innovatively revealed the mechanism of Cd-induced cell growth which offers a fresh perspective on preventing Cd toxicity through glutamine metabolism.
Collapse
Affiliation(s)
- Yanqiu Yang
- Department of Occupational and Environmental Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
| | - Chunpeng Gao
- Multi-Disciplinary Treatment (MDT) office, Dalian Municipal Central Hospital, Dalian 116003, China
| | - Qiujuan Li
- Department of Occupational and Environmental Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
| | - Yong Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China.
| | - Jun Cao
- Department of Occupational and Environmental Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China.
| |
Collapse
|
3
|
Romashin D, Rusanov A, Tolstova T, Varshaver A, Netrusov A, Kozhin P, Luzgina N. Loss of mutant p53 in HaCaT keratinocytes promotes cadmium-induced keratin 17 expression and cell death. Biochem Biophys Res Commun 2024; 709:149834. [PMID: 38547608 DOI: 10.1016/j.bbrc.2024.149834] [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: 01/17/2024] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND Cadmium exposure induces dermatotoxicity and epidermal barrier disruption and leads to the development of various pathologies. HaCaT cells are immortalized human keratinocytes that are widely used as alternatives to primary human keratinocytes, particularly for evaluating cadmium toxicity. HaCaT cells bear two gain-of-function (GOF) mutations in the TP53 gene, which strongly affect p53 function. Mutant forms of p53 are known to correlate with increased resistance to various stimuli, including exposure to cytotoxic substances. In addition, keratin 17 (KRT17) was recently shown to be highly expressed in HaCaT cells in response to genotoxic stress. Moreover, p53 is a direct transcriptional repressor of KRT17. However, the impact of TP53 mutations in HaCaT cells on the regulation of cell death and keratin 17 expression is unclear. In this study, we aimed to evaluate the impact of p53 on the response to Cd-induced cytotoxicity. METHODS AND RESULTS Employing the MTT assay and Annexin V/propidium iodide staining, we demonstrated that knockout of TP53 leads to a decrease in the sensitivity of HaCaT cells to the cytotoxic effects of cadmium. Specifically, HaCaT cells with TP53 knockout (TP53 KO HaCaT) exhibited cell death at a cadmium concentration of 10 μM or higher, whereas wild-type cells displayed cell death at a concentration of 30 μM. Furthermore, apoptotic cells were consistently detected in TP53 KO HaCaT cells upon exposure to low concentrations of cadmium (10 and 20 μM) but not in wild-type cells. Our findings also indicate that cadmium cytotoxicity is mediated by reactive oxygen species (ROS), which were significantly increased only in TP53 knockout cells treated with 30 μM cadmium. An examination of proteomic data revealed that TP53 knockout in HaCaT cells resulted in the upregulation of proteins involved in the regulation of apoptosis, redox systems, and DNA repair. Moreover, RT‒qPCR and immunoblotting showed that cadmium toxicity leads to dose-dependent induction of keratin 17 in p53-deficient cells but not in wild-type cells. CONCLUSIONS The connection between mutant p53 in HaCaT keratinocytes and increased resistance to cadmium toxicity was demonstrated for the first time. Proteomic profiling revealed that TP53 knockout in HaCaT cells led to the activation of apoptosis regulatory circuits, redox systems, and DNA repair. In addition, our data support the involvement of keratin 17 in the regulation of DNA repair and cell death. Apparently, the induction of keratin 17 is p53-independent but may be inhibited by mutant p53.
Collapse
Affiliation(s)
- Daniil Romashin
- Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow, 119121, Russia
| | - Alexander Rusanov
- Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow, 119121, Russia.
| | - Tatiana Tolstova
- Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow, 119121, Russia
| | - Alexandra Varshaver
- Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow, 119121, Russia
| | - Alexander Netrusov
- Lomonosov Moscow State University, GSP-1, Leninskie Gory, Moscow, 119991, Russia
| | - Peter Kozhin
- Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow, 119121, Russia
| | - Nataliya Luzgina
- Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow, 119121, Russia
| |
Collapse
|
4
|
Tu Z, Tang L, Khan FU, Hu M, Shen H, Wang Y. Low-frequency noise aggravates the toxicity of cadmium in sea slug Onchdium reevesii. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:169558. [PMID: 38135081 DOI: 10.1016/j.scitotenv.2023.169558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/25/2023] [Accepted: 12/18/2023] [Indexed: 12/24/2023]
Abstract
Industrial development not only triggers heavy metal pollution but also introduces a less easily discernible disturbance: low-frequency noise pollution. Low-frequency noise can disrupt wildlife behavior, potentially exerting complex effects through interacting with heavy metals. Nevertheless, the cumulative impacts of low-frequency noise and cadmium (Cd) pollution on marine organisms remain largely unexplored. This study aimed to evaluate the immune defense response of sea slugs (Onchdium reevesii) exposed to Cd (1.32 mg/L) and low-frequency noise (500 Hz, 1000 Hz). Our results show that Cd exposure results in Cd2+ accumulation in the sea slug's hepatopancreas, leading to a decrease in total antioxidant capacity (TAC) and a significant increase in enzyme activities, including glutathione (GSH), lipid peroxidation (LPO), and aspartate transferase (AST). Additionally, there is a substantial upregulation in the expression of genes related to tumor protein p53 (p53), Cytochrome C (CytC), Caspase 3, and Caspase 9, as well as metallothionein (MT) and heat shock protein 70 (Hsp70) genes. Concurrently, an excessive production of reactive oxygen species (ROS) occurs in the hemocytes, resulting in apoptosis and subsequent diminished cell viability, with these effects positively correlating with the exposure duration. Furthermore, when sea slugs were exposed to both Cd and low-frequency noise, there was a decrease in the hepatopancreas's antioxidant capacity and an enhancement in hemocytes immune responses, which positively correlated with low-frequency noise frequency. The comprehensive assessment of biomarker responses highlights that low-frequency noise has the potential to amplify the deleterious effects of Cd on sea slug physiology, with this negative impact positively linked to noise frequency. Consequently, our study underscores that the combined influence of low-frequency noise and Cd pollution magnifies the effects on sea slug health. This could potentially disrupt the population stability of this species within its natural habitat, providing fresh insights into the evaluation of cumulative environmental pollution risks.
Collapse
Affiliation(s)
- Zhihan Tu
- International Research Center for Marine Biosciences, Shanghai Ocean University, Ministry of Science and Technology, College of Fisheries and Life Science, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
| | - Liusiqiao Tang
- International Research Center for Marine Biosciences, Shanghai Ocean University, Ministry of Science and Technology, College of Fisheries and Life Science, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
| | - Fahim Ullah Khan
- International Research Center for Marine Biosciences, Shanghai Ocean University, Ministry of Science and Technology, College of Fisheries and Life Science, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
| | - Menghong Hu
- International Research Center for Marine Biosciences, Shanghai Ocean University, Ministry of Science and Technology, College of Fisheries and Life Science, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
| | - Heding Shen
- International Research Center for Marine Biosciences, Shanghai Ocean University, Ministry of Science and Technology, College of Fisheries and Life Science, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China.
| | - Youji Wang
- International Research Center for Marine Biosciences, Shanghai Ocean University, Ministry of Science and Technology, College of Fisheries and Life Science, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
5
|
Johnson L, Sarosiek KA. Role of intrinsic apoptosis in environmental exposure health outcomes. Trends Mol Med 2024; 30:56-73. [PMID: 38057226 DOI: 10.1016/j.molmed.2023.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/31/2023] [Accepted: 11/06/2023] [Indexed: 12/08/2023]
Abstract
Environmental exposures are linked to diseases of high public health concern, including cancer, neurodegenerative disorders, and autoimmunity. These diseases are caused by excessive or insufficient cell death, prompting investigation of mechanistic links between environmental toxicants and dysregulation of cell death pathways, including apoptosis. This review describes how legacy and emerging environmental exposures target the intrinsic apoptosis pathway to potentially drive pathogenesis. Recent discoveries reveal that dynamic regulation of apoptosis may heighten the vulnerability of healthy tissues to exposures in children, and that apoptotic signaling can guide immune responses, tissue repair, and tumorigenesis. Understanding how environmental toxicants dysregulate apoptosis will uncover opportunities to deploy apoptosis-modulating agents for the treatment or prevention of exposure-linked diseases.
Collapse
Affiliation(s)
- Lissah Johnson
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Molecular and Integrative Physiological Sciences Program, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Laboratory for Systems Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Kristopher A Sarosiek
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Molecular and Integrative Physiological Sciences Program, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Laboratory for Systems Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Cancer Center, Boston, MA, USA.
| |
Collapse
|
6
|
Hyun M, Kim H, Kim J, Lee J, Lee HJ, Rathor L, Meier J, Larner A, Lee SM, Moon Y, Choi J, Han SM, Heo JD. Melatonin protects against cadmium-induced oxidative stress via mitochondrial STAT3 signaling in human prostate stromal cells. Commun Biol 2023; 6:157. [PMID: 36750754 PMCID: PMC9905543 DOI: 10.1038/s42003-023-04533-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 01/26/2023] [Indexed: 02/09/2023] Open
Abstract
Melatonin protects against Cadmium (Cd)-induced toxicity, a ubiquitous environmental toxicant that causes adverse health effects by increasing reactive oxygen species (ROS) production and mitochondrial dysfunction. However, the underlying mechanism remains unclear. Here, we demonstrate that Cd exposure reduces the levels of mitochondrially-localized signal transducer and activator of transcription 3 (mitoSTAT3) using human prostate stromal cells and mouse embryonic fibroblasts. Melatonin enhances mitoSTAT3 abundance following Cd exposure, which is required to attenuate ROS damage, mitochondrial dysfunction, and cell death caused by Cd exposure. Moreover, melatonin increases mitochondrial levels of GRIM-19, an electron transport chain component that mediates STAT3 import into mitochondria, which are downregulated by Cd. In vivo, melatonin reverses the reduced size of mouse prostate tissue and levels of mitoSTAT3 and GRIM-19 induced by Cd exposure. Together, these data suggest that melatonin regulates mitoSTAT3 function to prevent Cd-induced cytotoxicity and could preserve mitochondrial function during Cd-induced stress.
Collapse
Affiliation(s)
- Moonjung Hyun
- grid.418982.e0000 0004 5345 5340Gyeongnam Biohealth Research Center, Gyeongnam Branch Institute, Korea Institute of Toxicology, Jinju, 52834 Republic of Korea
| | - Hyejin Kim
- grid.418982.e0000 0004 5345 5340Gyeongnam Biohealth Research Center, Gyeongnam Branch Institute, Korea Institute of Toxicology, Jinju, 52834 Republic of Korea
| | - Jehein Kim
- grid.418982.e0000 0004 5345 5340Gyeongnam Biohealth Research Center, Gyeongnam Branch Institute, Korea Institute of Toxicology, Jinju, 52834 Republic of Korea
| | - Juhong Lee
- grid.418982.e0000 0004 5345 5340Gyeongnam Biohealth Research Center, Gyeongnam Branch Institute, Korea Institute of Toxicology, Jinju, 52834 Republic of Korea
| | - Ho Jeong Lee
- grid.418982.e0000 0004 5345 5340Gyeongnam Biohealth Research Center, Gyeongnam Branch Institute, Korea Institute of Toxicology, Jinju, 52834 Republic of Korea
| | - Laxmi Rathor
- grid.15276.370000 0004 1936 8091Department of Physiology and Aging, College of Medicine, Institute on Aging, University of Florida, Gainesville, FL USA
| | - Jeremy Meier
- grid.410711.20000 0001 1034 1720Division of Hematology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC USA
| | - Andrew Larner
- grid.224260.00000 0004 0458 8737Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA USA
| | - Seon Min Lee
- grid.418982.e0000 0004 5345 5340Gyeongnam Biohealth Research Center, Gyeongnam Branch Institute, Korea Institute of Toxicology, Jinju, 52834 Republic of Korea
| | - Yeongyu Moon
- grid.418982.e0000 0004 5345 5340Gyeongnam Biohealth Research Center, Gyeongnam Branch Institute, Korea Institute of Toxicology, Jinju, 52834 Republic of Korea
| | - Jungil Choi
- grid.418982.e0000 0004 5345 5340Gyeongnam Biohealth Research Center, Gyeongnam Branch Institute, Korea Institute of Toxicology, Jinju, 52834 Republic of Korea
| | - Sung Min Han
- Department of Physiology and Aging, College of Medicine, Institute on Aging, University of Florida, Gainesville, FL, USA.
| | - Jeong-Doo Heo
- Gyeongnam Biohealth Research Center, Gyeongnam Branch Institute, Korea Institute of Toxicology, Jinju, 52834, Republic of Korea.
| |
Collapse
|
7
|
Burke AJ, McAuliffe JD, Natoni A, Ridge S, Sullivan FJ, Glynn SA. Chronic nitric oxide exposure induces prostate cell carcinogenesis, involving genetic instability and a pro-tumorigenic secretory phenotype. Nitric Oxide 2022; 127:44-53. [PMID: 35872082 DOI: 10.1016/j.niox.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/12/2022] [Accepted: 07/15/2022] [Indexed: 11/26/2022]
Abstract
Prostate cancer is a leading cause of cancer death in men. Inflammation and overexpression of inducible nitric oxide synthase (NOS2) have been implicated in prostate carcinogenesis. We aimed to explore the hypothesis that nitric oxide NO exerts pro-tumorigenic effects on prostate cells at physiologically relevant levels contributing to carcinogenesis. We investigated the impact of acute exposure of normal immortalised prostate cells (RWPE-1) to NO on cell proliferation and activation of DNA damage repair pathways. Furthermore we investigated the long term effects of chronic NO exposure on RWPE-1 cell migration and invasion potential and hallmarks of transformation. Our results demonstrate that NO induces DNA damage as indicated by γH2AX foci and p53 activation resulting in a G1/S phase block and activation of 53BP1 DNA damage repair protein. Long term adaption to NO results in increased migration and invasion potential, acquisition of anchorage independent growth and increased resistance to chemotherapy. This was recapitulated in PC3 and DU145 prostate cancer cells which upon chronic exposure to NO displayed increased cell migration, colony formation and increased resistance to chemotherapeutics. These findings indicate that NO may play a key role in the development of prostate cancer and the acquisition of an aggressive metastatic phenotype.
Collapse
Affiliation(s)
- Amy J Burke
- Prostate Cancer Institute, School of Medicine, National University of Ireland Galway, Galway, H91 TK33, Ireland
| | - Jake D McAuliffe
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway, Galway, H91 TK33, Ireland
| | - Alessandro Natoni
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Sarah Ridge
- Prostate Cancer Institute, School of Medicine, National University of Ireland Galway, Galway, H91 TK33, Ireland
| | - Francis J Sullivan
- Prostate Cancer Institute, School of Medicine, National University of Ireland Galway, Galway, H91 TK33, Ireland
| | - Sharon A Glynn
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway, Galway, H91 TK33, Ireland.
| |
Collapse
|
8
|
The role of autophagy in cadmium-induced acute toxicity in glomerular mesangial cells and tracking polyubiquitination of cytoplasmic p53 as a biomarker. Exp Mol Med 2022; 54:685-696. [PMID: 35624155 PMCID: PMC9166781 DOI: 10.1038/s12276-022-00782-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 04/12/2022] [Accepted: 04/19/2022] [Indexed: 11/21/2022] Open
Abstract
Cadmium (Cd) is a highly toxic environmental pollutant that can severely damage the kidneys. Here, we show that Cd-induced apoptosis is promoted by the cytoplasmic polyubiquitination of p53 (polyUb-p53), which is regulated by the polyubiquitination of SQSTM1/p62 (polyUb-p62) and autophagy in mouse kidney mesangial cells (MES13E cells). p53 was detected in monomeric and different high-molecular-weight (HMW) forms after Cd exposure. Monomeric p53 levels decreased in a concentration- and time-dependent manner. HMW-p53 transiently accumulated in the cytoplasm independent of proteasome inhibition. The expression patterns of p53 were similar to those of p62 upon Cd exposure, and the interactions between polyUb-p53 and polyUb-p62 were observed using immunoprecipitation. P62 knockdown reduced polyUb-p53 and upregulated nuclear monomeric p53, whereas p53 knockdown reduced polyUb-p62. Autophagy inhibition induced by ATG5 knockdown reduced Cd-induced polyUb-p62 and polyUb-p53 but upregulated the levels of nuclear p53. Pharmacological inhibition of autophagy by bafilomycin A1 increased polyUb-p62 and polyUb-p53 in the cytoplasm, indicating that p53 protein levels and subcellular localization were regulated by polyUb-p62 and autophagy. Immunoprecipitation and immunofluorescence revealed an interaction between p53 and LC3B, indicating that p53 was taken up by autophagosomes. Cd-resistant RMES13E cells and kidney tissues from mice continuously injected with Cd had reduced polyUb-p53, polyUb-p62, and autophagy levels. Similar results were observed in renal cell carcinoma cell lines. These results indicate that cytoplasmic polyUb-p53 is a potential biomarker for Cd-induced acute toxicity in mesangial cells. In addition, upregulation of nuclear p53 may protect cells against Cd cytotoxicity, but abnormal p53 accumulation may contribute to tumor development. The cellular localization and chemical modification of a protein that acts as a critical safeguard against cellular damage may directly contribute to the toxic effects of cadmium. P53 is an essential tumor suppressor that is also involved in numerous other important biological functions. Ki-Tae Jung and Seon-Hee Oh of Chosun University, Gwangju, South Korea have now demonstrated that this protein also undergoes rapid changes in response to the environmental pollutant cadmium. P53 normally manages gene expression in the nucleus, but the authors found that it is rapidly shuttled to the cytoplasm and subjected to extensive chemical modification in cadmium-treated cultured kidney cells. This relocation appears to contribute directly to subsequent cell death, and the authors suggest that this P53 response could be an important biomarker for diagnosing human cadmium exposure.![]()
Collapse
|
9
|
Famurewa AC, Renu K, Eladl MA, Chakraborty R, Myakala H, El-Sherbiny M, Elsherbini DMA, Vellingiri B, Madhyastha H, Ramesh Wanjari U, Goutam Mukherjee A, Valsala Gopalakrishnan A. Hesperidin and hesperetin against heavy metal toxicity: Insight on the molecular mechanism of mitigation. Biomed Pharmacother 2022; 149:112914. [DOI: 10.1016/j.biopha.2022.112914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 11/02/2022] Open
|
10
|
Chu CQ. Highlights of Strategies Targeting Fibroblasts for Novel Therapies for Rheumatoid Arthritis. Front Med (Lausanne) 2022; 9:846300. [PMID: 35252279 PMCID: PMC8891528 DOI: 10.3389/fmed.2022.846300] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 01/26/2022] [Indexed: 12/11/2022] Open
Abstract
Synovial fibroblasts of rheumatoid arthritis (RA) play a critical role in perpetuation of chronic inflammation by interaction with immune and inflammatory cells and in cartilage and bone invasion, but current therapies for RA are not directly targeted fibroblasts. Selectively fibroblast targeted therapy has been hampered because of lack of fibroblast specific molecular signature. Recent advancement in technology enabled us to gain insightful information concerning RA synovial fibroblast subpopulations and functions. Exploring fibroblast targeted therapies have been focused on inducing cell death via fibroblast associated proteins; interrupting fibroblast binding to matrix protein; blocking intercellular signaling between fibroblasts and endothelial cells; inhibiting fibroblast proliferation and invasion; promoting cell apoptosis and inducing cellular senescence, and modulating fibroblast glucose metabolism. Translation into clinical studies of these fibroblast targeted strategies is required for evaluation for their clinical application, in particular for combination therapy with current immune component targeted therapies. Here, several strategies of fibroblast targeted therapy are highlighted.
Collapse
|
11
|
de Almeida Rodrigues P, Ferrari RG, Kato LS, Hauser-Davis RA, Conte-Junior CA. A Systematic Review on Metal Dynamics and Marine Toxicity Risk Assessment Using Crustaceans as Bioindicators. Biol Trace Elem Res 2022; 200:881-903. [PMID: 33788164 DOI: 10.1007/s12011-021-02685-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/18/2021] [Indexed: 12/13/2022]
Abstract
Metals, many of which are potentially toxic, are present in the aquatic environment originated from both natural and anthropogenic sources. In these ecosystems, these elements are mostly deposited in the sediment, followed by water dissolution, potentially contaminating resident biota. Among several aquatic animals, crustaceans are considered excellent bioindicators, as they live in close contact with contaminated sediment. The accumulation of metal, whether they are classified as essential, when in excessive quantities or nonessential, not only cause damage to the health of these animals, but also to the man who consumes seafood. Among the main toxic elements to animal and human health are aluminum, arsenic, cadmium, chromium, copper, lead, mercury, nickel and silver. In this context, this systematic review aimed to investigate the dynamics of these metals in water, the main bioaccumulative tissues in crustaceans, the effects of these contaminants on animal and human health, and the regulatory limits for these metals worldwide. A total of 91 articles were selected for this review, and an additional 68 articles not found in the three assessed databases were considered essential and included, totaling 159 articles published between 2010 and 2020. Our results indicate that both chemical speciation and abiotic factors such as pH, oxygen and salinity in aquatic environments affect element bioavailability, dynamics, and toxicity. Among crustaceans, crabs are considered the main bioindicator biological system, with the hepatopancreas appearing as the main bioaccumulator organ. Studies indicate that exposure to these elements may result in nervous, respiratory, and reproductive system effects in both animals and humans. Finally, many studies indicate that the concentrations of these elements in crustaceans intended for human consumption exceed limits established by international organizations, both with regard to seafood metal contents and well as daily, weekly, or monthly intake limits set for humans, indicating consumer health risks.
Collapse
Affiliation(s)
- Paloma de Almeida Rodrigues
- Molecular and Analytical Laboratory Center, Department of Food Technology, Faculty of Veterinary, Universidade Federal Fluminense, Niterói, 24230-340, Brazil
| | - Rafaela Gomes Ferrari
- Chemistry Institute, Department of Biochemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-909, Brazil.
- Agrarian Sciences Center, Department of Zootechnics, Federal University of Paraiba, Paraiba, Brazil.
| | - Lilian Seiko Kato
- Chemistry Institute, Department of Biochemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-909, Brazil
| | - Rachel Ann Hauser-Davis
- Laboratório de Avaliação e Promoção da Saúde Ambiental, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, 21040-360, Brazil
| | - Carlos Adam Conte-Junior
- Molecular and Analytical Laboratory Center, Department of Food Technology, Faculty of Veterinary, Universidade Federal Fluminense, Niterói, 24230-340, Brazil
- Chemistry Institute, Department of Biochemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-909, Brazil
- National Institute of Health Quality Control, Fundação Oswaldo Cruz, Rio de Janeiro, 21040-900, Brazil
| |
Collapse
|
12
|
Kar I, Patra AK. Tissue Bioaccumulation and Toxicopathological Effects of Cadmium and Its Dietary Amelioration in Poultry-a Review. Biol Trace Elem Res 2021; 199:3846-3868. [PMID: 33405085 DOI: 10.1007/s12011-020-02503-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/17/2020] [Indexed: 12/20/2022]
Abstract
Cadmium (Cd) has been recognized as one of the most toxic heavy metals, which is continuously discharged into environments through anthropogenic (industrial activities, fertilizer production, and waste disposal) and natural sources with anthropogenic sources contributing greater than the natural sources. Therefore, Cd concentration sometimes increases in feeds, fodders, water bodies, and tissues of livestock including poultry in the vicinity of the industrial areas, which causes metabolic, structural, and functional changes of different organs of all animals. In poultry, bioaccumulation of Cd occurs in several organs mainly in the liver, kidney, lung, and reproductive organs due to its continuous exposure. Intake of Cd reduces growth and egg laying performance and feed conversion efficiency in poultry. Chronic exposure of Cd at low doses can also alter the microscopic structures of tissues, particularly in the liver, kidney, brain, pancreas, intestine, and reproductive organs due to increased content of Cd in these tissues. Continuous Cd exposure causes increased oxidative stress at cellular levels due to over-production of reactive oxygen species, exhausting antioxidant defense mechanisms. This leads to disruption of biologically relevant molecules, particularly nucleic acid, protein and lipid, and subsequently apoptosis, cell damage, and necrotic cell death. The histopatholocal changes in the liver, kidneys, and other organs are adversely reflected in hemogram and serum biochemical and enzyme activities. The present review discusses about Cd bioaccumulation and histopathological alterations in different tissues, pathogenesis of Cd toxicity, blood-biochemical changes, and its different ameliorative measures in poultry.
Collapse
Affiliation(s)
- Indrajit Kar
- Department of Avian Sciences, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, 700037, India
| | - Amlan Kumar Patra
- Department of Animal Nutrition, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, 700037, India.
| |
Collapse
|
13
|
Patriarca EJ, Cermola F, D’Aniello C, Fico A, Guardiola O, De Cesare D, Minchiotti G. The Multifaceted Roles of Proline in Cell Behavior. Front Cell Dev Biol 2021; 9:728576. [PMID: 34458276 PMCID: PMC8397452 DOI: 10.3389/fcell.2021.728576] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Herein, we review the multifaceted roles of proline in cell biology. This peculiar cyclic imino acid is: (i) A main precursor of extracellular collagens (the most abundant human proteins), antimicrobial peptides (involved in innate immunity), salivary proteins (astringency, teeth health) and cornifins (skin permeability); (ii) an energy source for pathogenic bacteria, protozoan parasites, and metastatic cancer cells, which engage in extracellular-protein degradation to invade their host; (iii) an antistress molecule (an osmolyte and chemical chaperone) helpful against various potential harms (UV radiation, drought/salinity, heavy metals, reactive oxygen species); (iv) a neural metabotoxin associated with schizophrenia; (v) a modulator of cell signaling pathways such as the amino acid stress response and extracellular signal-related kinase pathway; (vi) an epigenetic modifier able to promote DNA and histone hypermethylation; (vii) an inducer of proliferation of stem and tumor cells; and (viii) a modulator of cell morphology and migration/invasiveness. We highlight how proline metabolism impacts beneficial tissue regeneration, but also contributes to the progression of devastating pathologies such as fibrosis and metastatic cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gabriella Minchiotti
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics “A. Buzzati Traverso”, Consiglio Nazionale delle Ricerche, Naples, Italy
| |
Collapse
|
14
|
Bimonte VM, Besharat ZM, Antonioni A, Cella V, Lenzi A, Ferretti E, Migliaccio S. The endocrine disruptor cadmium: a new player in the pathophysiology of metabolic diseases. J Endocrinol Invest 2021; 44:1363-1377. [PMID: 33501614 DOI: 10.1007/s40618-021-01502-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 01/05/2021] [Indexed: 12/13/2022]
Abstract
Cadmium (Cd), a highly toxic heavy metal, is found in soil, environment and contaminated water and food. Moreover, Cd is used in various industrial activities, such as electroplating, batteries production, fertilizers, while an important non-occupational source is represented by cigarette smoking, as Cd deposits in tobacco leaves. Since many years it is clear a strong correlation between Cd body accumulation and incidence of many diseases. Indeed, acute exposure to Cd can cause inflammation and affect many organs such as kidneys and liver. Furthermore, the attention has focused on its activity as environmental pollutant and endocrine disruptor able to interfere with metabolic and energy balance of living beings. Both in vitro and in vivo experiments have demonstrated that the Cd-exposure is related to metabolic diseases such as obesity, diabetes and osteoporosis even if human studies are still controversial. Recent data show that Cd-exposure is associated with atherosclerosis, hypertension and endothelial damage that are responsible for cardiovascular diseases. Due to the large environmental diffusion of Cd, in this review, we summarize the current knowledge concerning the role of Cd in the incidence of metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- V M Bimonte
- Department of Movement, Human and Health Sciences, Section of Health Sciences, Foro Italico University, Piazza Lauro De Bosis 6, 00195, Rome, Italy
| | - Z M Besharat
- Department of Experimental Medicine, Section of Medical Pathophysiology, Endocrinology and Food Sciences, Sapienza University of Rome, Viiale Regina Elena 324, 00161, Rome, Italy
| | - A Antonioni
- Department of Experimental Medicine, Section of Medical Pathophysiology, Endocrinology and Food Sciences, Sapienza University of Rome, Viiale Regina Elena 324, 00161, Rome, Italy
| | - V Cella
- Department of Movement, Human and Health Sciences, Section of Health Sciences, Foro Italico University, Piazza Lauro De Bosis 6, 00195, Rome, Italy
| | - A Lenzi
- Department of Experimental Medicine, Section of Medical Pathophysiology, Endocrinology and Food Sciences, Sapienza University of Rome, Viiale Regina Elena 324, 00161, Rome, Italy
| | - E Ferretti
- Department of Experimental Medicine, Section of Medical Pathophysiology, Endocrinology and Food Sciences, Sapienza University of Rome, Viiale Regina Elena 324, 00161, Rome, Italy
| | - S Migliaccio
- Department of Movement, Human and Health Sciences, Section of Health Sciences, Foro Italico University, Piazza Lauro De Bosis 6, 00195, Rome, Italy.
| |
Collapse
|
15
|
Acinetobacter tandoii ZM06 Assists Glutamicibacter nicotianae ZM05 in Resisting Cadmium Pressure to Preserve Dipropyl Phthalate Biodegradation. Microorganisms 2021; 9:microorganisms9071417. [PMID: 34209156 PMCID: PMC8307640 DOI: 10.3390/microorganisms9071417] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/23/2021] [Accepted: 06/28/2021] [Indexed: 12/13/2022] Open
Abstract
Dipropyl phthalate (DPrP) coexists with cadmium as cocontaminants in environmental media. A coculture system including the DPrP-degrading bacterium Glutamicibacter nicotianae ZM05 and the nondegrading bacterium Acinetobacter tandoii ZM06 was artificially established to degrade DPrP under Cd(II) stress. Strain ZM06 relieved the pressure of cadmium on strain ZM05 and accelerated DPrP degradation in the following three ways: first, strain ZM06 adsorbed Cd(II) on the cell surface (as observed by scanning electron microscopy) to decrease the concentration of Cd(II) in the coculture system; second, the downstream metabolites of ZM05 were utilized by strain ZM06 to reduce metabolite inhibition; and third, strain ZM06 supplied amino acids and fatty acids to strain ZM05 to relieve stress during DPrP degradation, which was demonstrated by comparative transcriptomic analysis. This study provides an elementary understanding of how microbial consortia improve the degradation efficiency of organic pollutants under heavy metals contamination.
Collapse
|
16
|
Cui ZG, Ahmed K, Zaidi SF, Muhammad JS. Ins and outs of cadmium-induced carcinogenesis: Mechanism and prevention. Cancer Treat Res Commun 2021; 27:100372. [PMID: 33865114 DOI: 10.1016/j.ctarc.2021.100372] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 04/03/2021] [Accepted: 04/06/2021] [Indexed: 11/16/2022]
Abstract
Cadmium (Cd) is a heavy metal and a highly toxic pollutant that is released into the environment as a byproduct of most modern factories and industries. Cd enters our body in significant quantities from contaminated water, cigarette smoke, or food product to many detrimental health hazards. Based on causal association all the Cd-related or derived compounds have been classified as carcinogens. In this study, we present an overview of the published literature to understand the molecular mechanisms for Cd-induced carcinogenesis and its prevention. In acute Cd poisoning production of reactive oxygen species is a key factor. However, chronic Cd exposure can transform cells to become more resistant to oxidative stress. Also, as an epigenetic mechanism Cd acts indirectly on DNA repair mechanisms via alteration of reactions upstream. Those transformed cells acquire resistance to apoptosis and deregulation of calcium homeostasis. Leading to uncontrolled carcinogenic cell proliferation and inherent DNA lesions. Flavonoids commonly found in plant foods have been shown to have a protective effect against Cd-induced carcinogenicity. A wide variety of tumorigenic mechanisms involved in chronic Cd exposure and the beneficial effects of flavonoids against Cd-induced carcinogenicity necessitate further investigations.
Collapse
Affiliation(s)
- Zheng-Guo Cui
- Graduate School of Medicine, Henan Polytechnic University, Jiaozuo 454000, China; Department of Environmental Health, University of Fukui School of Medical Science, 23-3 Matsuoka Shimoaizuki, Eiheiji, Fukui 910-1193 Japan
| | - Kanwal Ahmed
- Department of Basic Medical Sciences, College of Medicine, King Saud Bin Abdulaziz University of Health Sciences, Jeddah, Saudi Arabia
| | - Syed Faisal Zaidi
- Department of Basic Medical Sciences, College of Medicine, King Saud Bin Abdulaziz University of Health Sciences, Jeddah, Saudi Arabia
| | - Jibran Sualeh Muhammad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates.
| |
Collapse
|
17
|
Saran U, Tyagi A, Chandrasekaran B, Ankem MK, Damodaran C. The role of autophagy in metal-induced urogenital carcinogenesis. Semin Cancer Biol 2021; 76:247-257. [PMID: 33798723 DOI: 10.1016/j.semcancer.2021.03.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022]
Abstract
Environmental and/or occupational exposure to metals such as Arsenic (As), Cadmium (Cd), and Chromium (Cr) have been shown to induce carcinogenesis in various organs, including the urogenital system. However, the mechanisms responsible for metal-induced carcinogenesis remain elusive. We and others have shown that metals are potent inducers of autophagy, which has been suggested to be an adaptive stress response to allow metal-exposed cells to survive in hostile environments. Albeit few, recent experimental studies have shown that As and Cd promote tumorigenesis via autophagy and that inhibition of autophagic signaling suppressed metal-induced carcinogenesis. In light of the newly emerging role of autophagic involvement in metal-induced carcinogenesis, the present review focuses explicitly on the mechanistic role of autophagy and potential signaling pathways involved in As-, Cd-, and Cr-induced urogenital carcinogenesis.
Collapse
Affiliation(s)
- Uttara Saran
- Department of Urology, University of Louisville, Louisville, KY, United States
| | - Ashish Tyagi
- Department of Urology, University of Louisville, Louisville, KY, United States
| | | | - Murali K Ankem
- Department of Urology, University of Louisville, Louisville, KY, United States
| | - Chendil Damodaran
- Department of Urology, University of Louisville, Louisville, KY, United States; College of Pharmacy, Department of Pharmaceutical Sciences, Texas A&M, College Station, TX, United States.
| |
Collapse
|
18
|
Xia X, Wu S, Zhou Z, Wang G. Microbial Cd(II) and Cr(VI) resistance mechanisms and application in bioremediation. JOURNAL OF HAZARDOUS MATERIALS 2021; 401:123685. [PMID: 33113721 DOI: 10.1016/j.jhazmat.2020.123685] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/16/2020] [Accepted: 08/05/2020] [Indexed: 05/21/2023]
Abstract
The heavy metals cadmium (Cd) and chromium (Cr) are extensively used in industry and result in water and soil contamination. The highly toxic Cd(II) and Cr(VI) are the most common soluble forms of Cd and Cr, respectively. They enter the human body through the food chain and drinking water and then cause serious illnesses. Microorganisms can adsorb metals or transform Cd(II) and Cr(VI) into insoluble or less bioavailable forms, and such strategies are applicable in Cd and Cr bioremediation. This review focuses on the highlighting of novel achievements on microbial Cd(II) and Cr(VI) resistance mechanisms and their bioremediation applications. In addition, the knowledge gaps and research perspectives are also discussed in order to build a bridge between the theoretical breakthrough and the resolution of Cd(II) and Cr(VI) contamination problems.
Collapse
Affiliation(s)
- Xian Xia
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China; Hubei Key Laboratory of Edible Wild Plants Conservation & Utilization, Hubei Engineering Research Center of Special Wild Vegetables Breeding and Comprehensive Utilization Technology, National Experimental Teaching Demonstrating Center, College of Life Sciences, Hubei Normal University, Huangshi, 435002, PR China
| | - Shijuan Wu
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Zijie Zhou
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Gejiao Wang
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China.
| |
Collapse
|
19
|
Kwok ML, Meng Q, Hu XL, Chung CT, Chan KM. Whole-transcriptome sequencing (RNA-seq) study of the ZFL zebrafish liver cell line after acute exposure to Cd 2+ ions. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2020; 228:105628. [PMID: 32971353 DOI: 10.1016/j.aquatox.2020.105628] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/03/2020] [Accepted: 09/06/2020] [Indexed: 06/11/2023]
Abstract
Cadmium (Cd) is a non-essential metal with no known biological function and a broad range of toxic effects in biological systems. We used whole-transcriptome sequencing (RNA-seq) to study the effects of Cd2+ toxicity in zebrafish liver cells, ZFL. The results of an RNA-Seq analysis of ZFL cells exposed to 5, 10 or 20 μM Cd2+ for 4- or 24-h. The differentially expressed genes affected by Cd2+ were analyzed by using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis to study the regulated pathways. Cd2+ regulated the expression of genes associated with cellular Cu, Zn, and Fe homeostasis, DNA replication leading to cell cycle arrest and apoptosis, and glutathione metabolism. Cd2+ boosted up the amino acid synthesis, possibly to support the glutathione metabolism for tackling the oxidative stress generated from Cd2+. Cd2+ stimulation was similar to heat or xenobiotics, based on the responses from ZFL such as endoplasmic reticulum stress and protein folding. We linked also those finding of gene activations relating to carcinogenesis of Cd. This paper provides a comprehensive analysis of the expression profiles induced by Cd2+ exposure in ZFL cells, as well as useful insights into the specific toxic effects.
Collapse
Affiliation(s)
- Man Long Kwok
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin., N.T., Hong Kong
| | - Qi Meng
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin., N.T., Hong Kong
| | - Xue Lei Hu
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin., N.T., Hong Kong
| | - Chun Ting Chung
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin., N.T., Hong Kong
| | - King Ming Chan
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin., N.T., Hong Kong.
| |
Collapse
|
20
|
Cadmium induces apoptosis via generating reactive oxygen species to activate mitochondrial p53 pathway in primary rat osteoblasts. Toxicology 2020; 446:152611. [PMID: 33031904 DOI: 10.1016/j.tox.2020.152611] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/27/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
Cadmium (Cd), a heavy metal produced by various industries, contaminates the environment and seriously damages the skeletal system of humans and animals. Recent studies have reported that Cd can affect the viability of cells, including osteoblasts, both in vivo and in vitro. However, the mechanism of Cd-induced apoptosis remains unclear. In the present study, primary rat osteoblasts were used to investigate the Cd-induced apoptotic mechanism. We found that treatment with 2 and 5 μM Cd for 12 h decreased osteoblast viability and increased apoptosis. Furthermore, Cd increased the generation of reactive oxygen species (ROS), and, thus, DNA damage measured via p-H2AX. The level of the nuclear transcription factor p53 was significantly increased, which upregulated the expression of PUMA, Noxa, Bax, and mitochondrial cytochrome c, downregulated the expression of Bcl-2, and increased the level of cleaved caspase-3. However, pretreatment with the ROS scavenger N-acetyl-l-cysteine (NAC) or the p53 transcription specific inhibitor PFT-α suppressed Cd-induced apoptosis. Our results indicate that Cd can induce apoptosis in osteoblasts by increasing the generation of ROS and activating the mitochondrial p53 signaling pathway, and this mechanism requires the transcriptional activation of p53.
Collapse
|
21
|
Roy A, Nethi SK, Suganya N, Raval M, Chatterjee S, Patra CR. Attenuation of cadmium-induced vascular toxicity by pro-angiogenic nanorods. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 115:111108. [DOI: 10.1016/j.msec.2020.111108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 05/07/2020] [Accepted: 05/19/2020] [Indexed: 01/03/2023]
|
22
|
Suljević D, Hodžić-Klapuh L, Handžić N, Fočak M. Morpho-functional alterations in lymphocytes and erythrocytes of Japanese quail due to prolonged in vivo exposure to heavy metal complexes. J Trace Elem Med Biol 2020; 59:126472. [PMID: 32088554 DOI: 10.1016/j.jtemb.2020.126472] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/20/2020] [Accepted: 02/03/2020] [Indexed: 10/25/2022]
Abstract
BACKGROUND Lead and cadmium are significant environmental pollutants that cause pathophysiological responses in many organs. Heavy metal absorption into many tissues is very fast due to a pronounced affinity for metallothioneins. METHOD Japanese quail were exposed to different concentrations of metals (cadmium 0.20 mg/L and lead 0.25 and 0.50 mg/L) for 20 days. Erythrocytes (normal and hemolyzed) and lymphocytes (normal and altered) were monitored in this study. The analysis observed the percentage of normal and altered cells, as well as erythrocyte surface area. Cell counts were analyzed using light microscopy, while surface area and cytological changes in cells and nuclei were analyzed using licensed software. RESULTS Different concentrations of metals have caused erythrocyte hemolysis as well as structural and morphological alterations in lymphocytes. Destruction of cell and nucleus membrane, changes in cell size, erythrocyte denucleation and reduced erythrocyte surface area were observed. Cadmium has caused erythrocyte hemolysis (29.30 %) and lymphocyte damage (92.10 %). Higher doses of lead resulted in greater damage to lymphocytes (63 %). Also, treatment with higher dose of lead produced a higher percentage of hemolyzed erythrocytes (19.20 %) in comparison to lower dose (9.90 %). CONCLUSION The toxicity of heavy metals leads to reduced maturation of the blast, which causes the appearance of immature cells in peripheral circulation and severe destruction of blood cell membranes. Erythrocyte hemolysis can lead to anemia, while lymphocyte damage can lead to lymphocytopenia.
Collapse
Affiliation(s)
- Damir Suljević
- All: Faculty of Science, Department of Biology, University of Sarajevo, Bosnia and Herzegovina.
| | - Lejla Hodžić-Klapuh
- All: Faculty of Science, Department of Biology, University of Sarajevo, Bosnia and Herzegovina.
| | - Nejira Handžić
- All: Faculty of Science, Department of Biology, University of Sarajevo, Bosnia and Herzegovina.
| | - Muhamed Fočak
- All: Faculty of Science, Department of Biology, University of Sarajevo, Bosnia and Herzegovina.
| |
Collapse
|
23
|
Shen R, Li Y, Yu L, Wu H, Cui R, Liu S, Song Y, Wang D. Ex vivo detection of cadmium-induced renal damage by using confocal Raman spectroscopy. JOURNAL OF BIOPHOTONICS 2019; 12:e201900157. [PMID: 31407491 DOI: 10.1002/jbio.201900157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 06/10/2023]
Abstract
Cadmium (Cd) is a toxic heavy metal which is harmful to environment and organisms. The reabsorption of Cd in kidney leads it to be the main damaged organ in animals under the Cd exposure. In this work, we applied confocal Raman spectroscopy to map the pathological changes in situ in normal and Cd-exposed mice kidney. The renal tissue from Cd-exposed group displayed a remarkable decreasing in the intensity of typical peaks related to mitochondria, DNA, proteins and lipids. On the contrary, the peaks of collagen in Cd-exposed group elevated significantly. The components in each tissue were identified and distinguished by principal component analysis. Furthermore, all the biological investigations in this study were consistent with the Raman spectrum detection, which revealed the progression and degree of lesion induced by Cd. The confocal Raman spectroscopy provides a new perspective for in situ monitoring of substances changes in tissues, which exhibits more comprehensive understanding of the pathogenic mechanisms of heavy metals in molecular toxicology.
Collapse
Affiliation(s)
- Rong Shen
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Yuee Li
- School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Linghui Yu
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Haining Wu
- School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Rong Cui
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Sha Liu
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Yanfeng Song
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Degui Wang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
24
|
Zimta AA, Schitcu V, Gurzau E, Stavaru C, Manda G, Szedlacsek S, Berindan-Neagoe I. Biological and molecular modifications induced by cadmium and arsenic during breast and prostate cancer development. ENVIRONMENTAL RESEARCH 2019; 178:108700. [PMID: 31520827 DOI: 10.1016/j.envres.2019.108700] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/07/2019] [Accepted: 08/26/2019] [Indexed: 06/10/2023]
Abstract
Breast and prostate cancer are two of the most common malignancies worldwide. Both cancers can develop into hormone -dependent or -independent subtypes and are associated to environmental exposure in the context of an inherited predisposition. As and Cd have been linked to the onset of both cancers, with the exception of As, which lacks a definitive association with breast carcinogenesis. The two elements exert an opposite effect dependent on acute versus chronic exposure. High doses of As or Cd were shown to induce cell death in acute experimental exposure, while chronic exposure triggers cell proliferation and viability, which is no longer limited by telomere shortening and apoptosis. The chronically exposed cells also increase their invasion capacity and tumorigenic potential. At molecular level, malignant transformation is evidenced mainly by up-regulation of BCL-2, MMP-2, MMP-9, VIM, Snail, Twist, MT, MLH and down-regulation of Casp-3, PTEN, E-CAD, and BAX. The signaling pathways most commonly activated are KRAS, p53, TGF-β, TNF-α, WNT, NRF2 and AKT. This knowledge could potentially raise public awareness over the health risks faced by the human population living or working in a polluted environment and smokers. Human exposure to As and Cd should be minimize as much as possible. Healthcare policies targeting people belonging to these risk categories should include analysis of: DNA damage, oxidative stress, molecular alterations, and systemic level of heavy metals and of essential minerals. In this review, we present the literature regarding cellular and molecular alterations caused by exposure to As or Cd, focusing on the malignant transformation of normal epithelial cells after long-term intoxication with these two carcinogens.
Collapse
Affiliation(s)
- Alina-Andreea Zimta
- MEDFUTURE - Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, 23 Gheorghe Marinescu Street, 400337, Cluj-Napoca, Romania
| | - Vlad Schitcu
- The Oncology Institute "Prof. Dr. Ion Chiricuta", Republicii 34-36 Street, 400015, Cluj-Napoca, Romania; "Iuliu Hatieganu" University of Medicine and Pharmacy, 8 Victor Babes Street, 400012, Cluj-Napoca, Romania
| | - Eugen Gurzau
- Cluj School of Public Health, College of Political, Administrative and Communication Sciences, Babes-Bolyai University, 7 Pandurilor Street, Cluj-Napoca, Romania; Environmental Health Center, 58 Busuiocului Street, 400240, Cluj-Napoca, Romania; Faculty of Environmental Science and Engineering, Babes-Bolyai University, 30 Fantanele Street, Cluj- Napoca, Romania
| | - Crina Stavaru
- Cantacuzino National Institute of Research and Development for Microbiology, 103 Splaiul Independentei Street, Bucharest, 050096, Romania
| | - Gina Manda
- "Victor Babes" National Institute of Pathology, 99-101 Splaiul Independentei Street, 050096, Bucharest, Romania
| | - Stefan Szedlacsek
- Department of Enzymology, Institute of Biochemistry of the Romanian Academy, 296 Splaiul Independentei Street, Bucharest, 060031, Romania
| | - Ioana Berindan-Neagoe
- MEDFUTURE - Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, 23 Gheorghe Marinescu Street, 400337, Cluj-Napoca, Romania; Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Marinescu 23 Street, 400337, Cluj-Napoca, Romania; Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuta", Republicii 34-36 Street, Cluj-Napoca, Romania.
| |
Collapse
|
25
|
Zhang H, Reynolds M. Cadmium exposure in living organisms: A short review. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 678:761-767. [PMID: 31085492 DOI: 10.1016/j.scitotenv.2019.04.395] [Citation(s) in RCA: 264] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/26/2019] [Accepted: 04/26/2019] [Indexed: 05/20/2023]
Abstract
Cadmium (Cd) is a toxic heavy metal that accumulates in living systems. Exposure can occur occupationally or environmentally. Workers within the electroplating, battery production, and pigment industries are at the highest risk for exposure and have been reported to have increased levels of Cd in their blood and urine. Environmental exposure can be the result of anthropogenic activities or smoking. Cd has a long half-life and bio accumulates in plants, invertebrates, and vertebrates. The toxic effects following exposure include growth retardation and organ system toxicity, with kidney and liver toxicity most reported with in higher organisms. At the molecular level, Cd leads to the production of reactive oxygen species, DNA damage, and inhibition of DNA repair. This article gives a brief overview of the correlations between exposure to cadmium occupationally and environmentally and levels measured in blood and urine. It also examines the bioaccumulation of cadmium in aquatic invertebrates and vertebrates indicating that accumulation varies not only by location but also within and between various species.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Biology, Washington College, 300 Washington Ave., Chestertown, MD 21620, USA
| | - Mindy Reynolds
- Department of Biology, Washington College, 300 Washington Ave., Chestertown, MD 21620, USA.
| |
Collapse
|
26
|
Pérez Díaz MFF, Plateo Pignatari MG, Filippa VP, Mohamed FH, Marchevsky EJ, Gimenez MS, Ramirez DC. A soybean-based diet modulates cadmium-induced vascular apoptosis. J Trace Elem Med Biol 2019; 52:239-246. [PMID: 30732889 DOI: 10.1016/j.jtemb.2019.01.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/21/2018] [Accepted: 01/11/2019] [Indexed: 10/27/2022]
Abstract
Cadmium (Cd) exposure has been associated with an increased risk of cardiovascular diseases. The diet is a modifiable source of protecting or damaging factors that may affect this risk. Herein we tested the hypothesis that a soybean-based diet (SBD) protects the vascular wall of the aorta against Cd-induced pro-inflammatory and pro-apoptotic effects. To test this hypothesis, we fed male Wistar rats for 60 days with a casein-based diet (CBD) or an SBD. These animals were also exposed to tap-water without (CBD-Co/SBD-Co) or with 15(CBD-15Cd/SBD-15Cd) or 100 (CBD-100Cd/SBD-100Cd) ppm of Cd. Inflammatory parameters (mRNAs and/or proteins) were measured in thoracic aorta tissue. These included inducible and endothelial nitric oxide synthases, cyclooxygenase-2, intracellular-adhesion molecule-1, and vascular cell-adhesion molecule-1. As pro-apoptotic parameters, we measured Bax and Bcl-2 mRNA/protein, as well as TUNEL positive cells in the aorta tissue. Compared to CBD-Co, inflammatory and apoptosis markers increased in the aorta with the concentration of Cd in the drinking water. These effects were not observed in either SBD-15Cd or SBD-100Cd, which were similar to CBD-Co. Cd content in serum and in aortas from animals fed CBD-Co/SBD-15Cd or CBD-Co/SBD-100Cd were similar suggesting that, if any, the effect of SBD is not due to changes in Cd bioaccumulation, but due to secondary effects linked to the composition of the dietary soybean flour. Our findings are consistent with a protective effect of an SBD against Cd-induced inflammation and apoptosis in the thoracic aorta in a rat model.
Collapse
Affiliation(s)
- Matías F F Pérez Díaz
- Laboratory of Nutrition, Environment, and Metabolism, Multidisciplinary Institute of Biological Research-San Luis, CCT-San Luis, CONICET-National University of San Luis, San Luis, 5700 San Luis, Argentina
| | - Maria G Plateo Pignatari
- Laboratory of Nutrition, Environment, and Metabolism, Multidisciplinary Institute of Biological Research-San Luis, CCT-San Luis, CONICET-National University of San Luis, San Luis, 5700 San Luis, Argentina; Laboratory of Experimental and Translational Medicine, Multidisciplinary Institute of Biological Research-San Luis, CCT-San Luis, CONICET-National University of San Luis, San Luis, 5700 San Luis, Argentina
| | - Verónica P Filippa
- Laboratory of Histology, Multidisciplinary Institute of Biological Research-San Luis, CCT-San Luis, CONICET-National University of San Luis, San Luis, 5700 San Luis, Argentina
| | - Fabián H Mohamed
- Laboratory of Histology, Multidisciplinary Institute of Biological Research-San Luis, CCT-San Luis, CONICET-National University of San Luis, San Luis, 5700 San Luis, Argentina
| | - Eduardo J Marchevsky
- Laboratory of Analytical Chemistry, School of Chemistry, Biochemistry, and Pharmacy, National University of San Luis, San Luis, 5700 San Luis, Argentina
| | - María S Gimenez
- Laboratory of Nutrition, Environment, and Metabolism, Multidisciplinary Institute of Biological Research-San Luis, CCT-San Luis, CONICET-National University of San Luis, San Luis, 5700 San Luis, Argentina.
| | - Dario C Ramirez
- Laboratory of Experimental and Translational Medicine, Multidisciplinary Institute of Biological Research-San Luis, CCT-San Luis, CONICET-National University of San Luis, San Luis, 5700 San Luis, Argentina.
| |
Collapse
|
27
|
Lee JY, Tokumoto M, Satoh M. Novel Mechanisms of Cadmium-Induced Toxicity in Renal Cells. CURRENT TOPICS IN ENVIRONMENTAL HEALTH AND PREVENTIVE MEDICINE 2019. [DOI: 10.1007/978-981-13-3630-0_12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
28
|
Mugnano M, Memmolo P, Miccio L, Grilli S, Merola F, Calabuig A, Bramanti A, Mazzon E, Ferraro P. In vitro cytotoxicity evaluation of cadmium by label-free holographic microscopy. JOURNAL OF BIOPHOTONICS 2018; 11:e201800099. [PMID: 30079614 DOI: 10.1002/jbio.201800099] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 07/17/2018] [Accepted: 08/01/2018] [Indexed: 05/04/2023]
Abstract
Among all environmental pollutants, the toxic heavy metal cadmium is considered as a human carcinogen. Cadmium may induce cell death by apoptosis in various cell types, although the underlying mechanisms are still unclear. In this paper we show how a label-free digital holography (DH)-based technique is able to quantify the evolution of key biophysical parameters of cells during the exposure to cadmium for the first time. Murine embryonic fibroblasts NIH 3T3 are chosen here as cellular model for studying the cadmium effects. The results demonstrate that DH is able to retrieve the temporal evolution of different key parameters such as cell volume, projected area, cell thickness and dry mass, thus providing a full quantitative characterization of the cell physical behaviour during cadmium exposure. Our results show that the label-free character of the technique would allow biologists to perform systematic and reliable studies on cell death process induced by cadmium and we believe that more in general this can be easily extended to others heavy metals, thus avoiding the time-consuming, expensive and invasive label-based procedures used nowadays in the field. In fact, pollution by heavy metals is severe issue that needs rapid and reliable methods to be settled.
Collapse
Affiliation(s)
- Martina Mugnano
- Department of Physical Sciences and Technologies of Matter (DSFTM), CNR, Institute of Applied Science & Intelligent Systems (CNR-ISASI), Pozzuoli, Italy
| | - Pasquale Memmolo
- Department of Physical Sciences and Technologies of Matter (DSFTM), CNR, Institute of Applied Science & Intelligent Systems (CNR-ISASI), Pozzuoli, Italy
| | - Lisa Miccio
- Department of Physical Sciences and Technologies of Matter (DSFTM), CNR, Institute of Applied Science & Intelligent Systems (CNR-ISASI), Pozzuoli, Italy
| | - Simonetta Grilli
- Department of Physical Sciences and Technologies of Matter (DSFTM), CNR, Institute of Applied Science & Intelligent Systems (CNR-ISASI), Pozzuoli, Italy
| | - Francesco Merola
- Department of Physical Sciences and Technologies of Matter (DSFTM), CNR, Institute of Applied Science & Intelligent Systems (CNR-ISASI), Pozzuoli, Italy
| | - Alejandro Calabuig
- Department of Physical Sciences and Technologies of Matter (DSFTM), CNR, Institute of Applied Science & Intelligent Systems (CNR-ISASI), Pozzuoli, Italy
| | - Alessia Bramanti
- Department of Physical Sciences and Technologies of Matter (DSFTM), CNR, Institute of Applied Science & Intelligent Systems (CNR-ISASI), Pozzuoli, Italy
- Department of Physical Sciences and Technologies of Matter (DSFTM), IRCCS Centre for Neuroscience Bonino-Pulejo, Messina, Italy
| | - Emanuela Mazzon
- Department of Physical Sciences and Technologies of Matter (DSFTM), IRCCS Centre for Neuroscience Bonino-Pulejo, Messina, Italy
| | - Pietro Ferraro
- Department of Physical Sciences and Technologies of Matter (DSFTM), CNR, Institute of Applied Science & Intelligent Systems (CNR-ISASI), Pozzuoli, Italy
| |
Collapse
|
29
|
Vella V, Malaguarnera R, Lappano R, Maggiolini M, Belfiore A. Recent views of heavy metals as possible risk factors and potential preventive and therapeutic agents in prostate cancer. Mol Cell Endocrinol 2017; 457:57-72. [PMID: 27773847 DOI: 10.1016/j.mce.2016.10.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 10/19/2016] [Accepted: 10/19/2016] [Indexed: 11/19/2022]
Abstract
Prostate cancer is the most common cancer in men in many industrialized countries. A role for androgens in prostate tumor progression is well recognized, while estrogens may cooperate with androgens in prostate carcinogenesis. The incidence of prostate cancer is highly variable in the different countries, suggesting an important role of environmental factors. Heavy metals are common environmental contaminants and some of them are confirmed or suspected human carcinogens. Some metals are endowed with estrogenic and/or androgenic activities and may play a role as cancer risk factors through this mechanism. Moreover, prostate cancer may present alterations in the intracellular balance of trace metals, such as zinc and copper, which are involved in several regulatory proteins. Herein, we review the possible role of environmental heavy metals and of metal-dyshomeostasis in prostate cancer development and promotion as well as the potential use of some metals in the prevention and therapy of prostate cancer.
Collapse
Affiliation(s)
- Veronica Vella
- School of Human and Social Science, Motor Sciences, University "Kore" of Enna, Enna, Italy
| | - Roberta Malaguarnera
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Antonino Belfiore
- Endocrinology, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy.
| |
Collapse
|
30
|
Cadmium Exposure as a Putative Risk Factor for the Development of Pancreatic Cancer: Three Different Lines of Evidence. BIOMED RESEARCH INTERNATIONAL 2017; 2017:1981837. [PMID: 29349066 PMCID: PMC5733953 DOI: 10.1155/2017/1981837] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 10/31/2017] [Indexed: 12/11/2022]
Abstract
Although profoundly studied, etiology of pancreatic cancer (PC) is still rather scant. Exposure to cadmium (Cd), a ubiquitous metal associated with well-established toxic and carcinogenic properties, has been hypothesized to one putative cause of PC. Hence, we analyzed recently published observational studies, meta-analyses, and experimental animal and in vitro studies with the aim of summarizing the evidence of Cd involvement in PC development and describing the possible mechanisms. Consolidation of epidemiological data on PC and exposure to Cd indicated a significant association with an elevated risk of PC among general population exposed to Cd. Cadmium exposure of laboratory animals was showed to cause PC supporting the findings suggested by human studies. The concordance with human and animal studies is buttressed by in vitro studies, although in vitro data interpretation is problematic. In most instances, only significant effects are reported, and the concentrations of Cd are excessive, which would skew interpretation. Previous reports suggest that oxidative stress, apoptotic changes, and DNA cross-linking and hypermethylation are involved in Cd-mediated carcinogenesis. Undoubtedly, a significant amount of work is still needed to achieve a better understanding of the Cd involvement in pancreatic cancer which could facilitate prevention, diagnosis, and therapy of this fatal disease.
Collapse
|
31
|
Bonaventura P, Lamboux A, Albarède F, Miossec P. Regulatory effects of zinc on cadmium-induced cytotoxicity in chronic inflammation. PLoS One 2017; 12:e0180879. [PMID: 28742830 PMCID: PMC5526586 DOI: 10.1371/journal.pone.0180879] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 06/22/2017] [Indexed: 02/01/2023] Open
Abstract
OBJECTIVES Zinc (Zn) has major effects on immune system activation while Cadmium (Cd) has anti-inflammatory and anti-proliferative effects in several chronic inflammatory contexts. The aim of this work was to investigate by which mechanisms Zn could compete with Cd and eventually counteract its deleterious effects. Rheumatoid arthritis (RA) synoviocytes exposed to cytokines were used as a model of chronic inflammation; osteoarthritis (OA) synoviocytes were used as control. METHODS Cell/medium fractionation constants were analyzed for different metals by inductively-coupled-plasma mass-spectrometry by comparison to the 70Zn spike. Interleukin-17 (IL-17) and tumor necrosis factor-alpha (TNF-α) were used to mimic inflammation. Gene expression of ZIP-8 importer, metallothioneins-1 (MT-1s) and the ratio between metalloprotease-3 and the tissue inhibitor of metalloproteinases (MMP-3)/TIMP-1) were evaluated after pre-exposure to cytokines and Cd, with or without the addition of exogenous Zn (0.9 ppm). Cell viability was measured by neutral red assay and IL-6 production by ELISA. RESULTS Synoviocytes selectively absorbed and retained Cd in comparison to Zn. Metal import increased with IL-17/TNF-α exposure, through the enhanced ZIP-8 expression. Zn did not modify ZIP-8 expression, while Cd reduced it (p<0.05). Zn induced a reduction of Cd-induced MT-1s expression, in particular of MT-1X (3-fold), and subsequently the final intra-cellular content of Cd. By reducing Cd accumulation in cells, Zn reversed Cd anti-proliferative and anti-inflammatory effects but preserved the low MMP-3/TIMP-1 ratio induced by Cd, which was enhanced by inflammatory conditions. CONCLUSION Zinc counteracts the deleterious effect of Cd by reducing its import and accumulation in the cell, without the reactivation of destructive pathways such as MMPs.
Collapse
Affiliation(s)
- Paola Bonaventura
- Department of Immunology and Rheumatology, Immunogenomics and inflammation research Unit EA 4130, University of Lyon, Edouard Herriot Hospital, Lyon, France
| | - Aline Lamboux
- Geology Laboratory–Department of Earth Sciences, Ecole Normale Supérieure de Lyon and CNRS Lyon, Lyon, France
| | - Francis Albarède
- Geology Laboratory–Department of Earth Sciences, Ecole Normale Supérieure de Lyon and CNRS Lyon, Lyon, France
| | - Pierre Miossec
- Department of Immunology and Rheumatology, Immunogenomics and inflammation research Unit EA 4130, University of Lyon, Edouard Herriot Hospital, Lyon, France
- * E-mail:
| |
Collapse
|
32
|
Pal D, Suman S, Kolluru V, Sears S, Das TP, Alatassi H, Ankem MK, Freedman JH, Damodaran C. Inhibition of autophagy prevents cadmium-induced prostate carcinogenesis. Br J Cancer 2017; 117:56-64. [PMID: 28588318 PMCID: PMC5520206 DOI: 10.1038/bjc.2017.143] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 04/20/2017] [Accepted: 04/25/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cadmium, an established carcinogen, is a risk factor for prostate cancer. Induction of autophagy is a prerequisite for cadmium-induced transformation and metastasis. The ability of Psoralidin (Pso), a non-toxic, orally bioavailable compound to inhibit cadmium-induced autophagy to prevent prostate cancer was investigated. METHODS Psoralidin was studied using cadmium-transformed prostate epithelial cells (CTPE), which exhibit high proliferative, invasive and colony forming abilities. Gene and protein expression were evaluated by qPCR, western blot, immunohistochemistry and immunofluorescence. Xenograft models were used to study the chemopreventive effects in vivo. RESULTS Cadmium-transformed prostate epithelial cells were treated with Pso resulting in growth inhibition, without causing toxicity to normal prostate epithelial cells (RWPE-1). Psoralidin-treatment of CTPE cells inhibited the expression of Placenta Specific 8, a lysosomal protein essential for autophagosome and autolysosome fusion, which resulted in growth inhibition. Additionally, Pso treatment caused decreased expression of pro-survival signalling proteins, NFκB and Bcl2, and increased expression of apoptotic genes. In vivo, Pso effectively suppressed CTPE xenografts growth, without any observable toxicity. Tumours from Pso-treated animals showed decreased autophagic morphology, mesenchymal markers expression and increased epithelial protein expression. CONCLUSIONS These results confirm that inhibition of autophagy by Pso plays an important role in the chemoprevention of cadmium-induced prostate carcinogenesis.
Collapse
Affiliation(s)
- Deeksha Pal
- Department of Urology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Suman Suman
- Department of Urology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Venkatesh Kolluru
- Department of Urology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Sophia Sears
- Department of Pharmacology &Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Trinath P Das
- Department of Urology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Houda Alatassi
- Department of Pathology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Murali K Ankem
- Department of Urology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Jonathan H Freedman
- Department of Pharmacology &Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Chendil Damodaran
- Department of Urology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
33
|
Protective effect of low dose intra-articular cadmium on inflammation and joint destruction in arthritis. Sci Rep 2017; 7:2415. [PMID: 28546541 PMCID: PMC5445071 DOI: 10.1038/s41598-017-02611-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 04/06/2017] [Indexed: 11/09/2022] Open
Abstract
Synovium hyperplasia characterizes joint diseases, such as rheumatoid arthritis (RA). The cytotoxic effect of low-dose Cadmium (Cd) was tested in vitro and ex vivo on synoviocytes, the mesenchymal key effector cells of inflammation and proliferation in arthritis. The anti-inflammatory and anti-proliferative effects of Cd were tested in vivo by intra-articular injection in the adjuvant induced arthritis rat joints, where the clinical scores and the consequences of arthritis were evaluated. Cell death through apoptosis was highly induced by Cd in inflammatory synoviocytes (80% reduction of cell viability, p < 0.01). TNF plus IL-17 cytokine combination induced a two-fold increase of Cd cell content by enhancing the ZIP-8 importer and the MT-1 homeostasis regulator expression. Addition of Cd reduced IL-6 production in TNF plus IL-17-activated synoviocytes (up to 83%, p < 0.05) and in ex-vivo synovium biopsies (up to 94%, p < 0.01). Cd-injection in rat joints improved arthritis, reducing clinical scores (arthritic score reduced from 4 to 2, p < 0.01), inflammatory cell recruitment (up to 50%, p < 0.01) and protecting from bone/cartilage destruction. This proof of concept study is supported by the limited Cd spread in body reservoirs, with low-dose Cd providing a safe risk/benefit ratio, without toxic effects on other cell types and organs.
Collapse
|
34
|
Gao Y, Xu Y, Wu D, Yu F, Yang L, Yao Y, Liang Z, Lau ATY. Progressive silencing of the zinc transporter Zip8 (Slc39a8) in chronic cadmium-exposed lung epithelial cells. Acta Biochim Biophys Sin (Shanghai) 2017; 49:444-449. [PMID: 28338971 DOI: 10.1093/abbs/gmx022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 03/01/2017] [Indexed: 02/05/2023] Open
Abstract
Cadmium (Cd), a non-essential metal, stealthily enters the cells by utilizing the essential metal importing pathways. The zinc transporters Zip8, Zip14, and divalent metal transporter 1 (Dmt1) are now emerging as several important metal transporters involved in cellular Cd incorporation and their expressions have been shown to be down-regulated in several Cd-resistant (CdR) cell lines, however, the involvement of these transporters during the development of Cd-resistance in lung cells is unclear. In this study, we therefore check the expression of these metal transporters in our previously established rat lung epithelial cells (LECs) and show that the level of Zip8 is progressively silenced when LECs are adapted to increasing concentrations of CdCl2 (from 1 to 20 μM). Subsequent measurement of the cellular Cd content indicated that CdR LECs exhibit a marked decrease of Cd accumulation, possibly due to the loss of Zip8 expression. We investigate the possibility that epigenetic silencing of the Zip8 gene by DNA hypermethylation is involved in the down-regulation of Zip8 expression. CdR LECs show a higher mRNA level of DNA methyltransferase 3b (Dnmt3b) than parental cells. Treatment of CdR LECs with 5-aza-2'-deoxycytidine, an inhibitor of DNA methyltransferases, reverted the expression of Zip8 and sensitivity to Cd in these cells, indicating the critical role of Zip8 for Cd import. Taken together, our results demonstrate that the progressive silencing of Zip8 expression is involved in the acquisition of resistance against Cd in lung cells, representing an adaptive survival mechanism that resists Cd-induced cytotoxicity.
Collapse
Affiliation(s)
- Yangmin Gao
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| | - Yanming Xu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| | - Dandan Wu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| | - Feiyuan Yu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| | - Lei Yang
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| | - Yue Yao
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| | - Zhanling Liang
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| | - Andy T Y Lau
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
35
|
Xu Z, Jin X, Pan T, Liu T, Wan N, Li S. Antagonistic effects of selenium on cadmium-induced apoptosis by restoring the mitochondrial dynamic equilibrium and energy metabolism in chicken spleens. Oncotarget 2017; 8:52629-52641. [PMID: 28881757 PMCID: PMC5581056 DOI: 10.18632/oncotarget.17539] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 04/12/2017] [Indexed: 12/13/2022] Open
Abstract
The aim of this study was to investigate the mechanism of cadmium-induced apoptosis in chicken spleens and the antagonistic effects of selenium. We duplicated the selenium-cadmium interaction model and examined the expression of apoptosis-, immune-, mitochondrial dynamics- and energy metabolism-related genes. The results demonstrated that after treatment with cadmium, the frequency of apoptosis was significantly increased, and the morphological characteristics of apoptosis were observed. The expression of pro-apoptotic genes was increased, and that of anti-apoptotic genes was decreased. The mRNA levels of tumor necrosis factor-α and interlenkin-1β were observably increased, but the interlenkin-2 and interferon-γ levels were markedly decreased. Furthermore, the mRNA and protein levels of dynamin-related protein 1 and mitochondrial fission factor were significantly enhanced, whereas mitofusin 1, mitofusin 2, and optic atrophy 1 were markedly decreased. The expression of hexokinase 1, hexokinase 2, aconitase 2, lactate dehydrogenase A, lactate dehydrogenase B, succinatedehydrogenase B, pyruvate kinase and phosphofructokinase were also reduced. Selenium supplements remarkably attenuated cadmium-induced effects (p < 0.05). Based on the above results, conclude that the cadmium treatment promoted a mitochondrial dynamic imbalance and reduced energy metabolism, leading to apoptosis and immune dysfunction in chicken spleens, and selenium had an antagonistic effect on Cd-induced apoptosis.
Collapse
Affiliation(s)
- Zhe Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Xi Jin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Tingru Pan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Tianqi Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Na Wan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Shu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, P. R. China
| |
Collapse
|
36
|
Ravindran G, Chakrabarty D, Sarkar A. Cell specific stress responses of cadmium-induced cytotoxicity. Anim Cells Syst (Seoul) 2016; 21:23-30. [PMID: 30460048 DOI: 10.1080/19768354.2016.1267041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 11/02/2016] [Accepted: 11/14/2016] [Indexed: 01/23/2023] Open
Abstract
Cadmium is one of the age old toxic heavy metal, detrimental to the biological system. In this study, we explored the cellular and molecular mechanisms induced on exposure to different concentrations of cadmium chloride (CdCl2), on three different human cell lines with wild type p53, viz., A549, HEK293 and HCT116. We investigated whether the cellular responses followed, displayed any specific pattern related to their viability, mitochondrial respiration, DNA damage and apoptotic gene expression. All the cell lines showed decrease in viability following exposure to CdCl2. p53 was transcriptionally down regulated in all the three cell lines, but with different extents, in response to increasing concentration of cadmium. The cellular responses of the three cell lines were compared with that of a p53 knock out cell line (HCT116p53-/-). The p53 knock out cell line was highly sensitive to cadmium-induced toxicity; so was the cell line in which p53 mRNA expression was highly down regulated. This might implicate an unknown protective role of p53 signaling during heavy metal toxicity and that one of the possible mechanisms by which cadmium manifests its cytotoxic effect is through the transcriptional down regulation of p53 gene.
Collapse
Affiliation(s)
- Geethanjali Ravindran
- CMBL, Department of Biological Sciences, BITS Pilani K K Birla Goa Campus, Zuarinagar, Goa, India
| | - Dibakar Chakrabarty
- CMBL, Department of Biological Sciences, BITS Pilani K K Birla Goa Campus, Zuarinagar, Goa, India
| | - Angshuman Sarkar
- CMBL, Department of Biological Sciences, BITS Pilani K K Birla Goa Campus, Zuarinagar, Goa, India
| |
Collapse
|
37
|
Transcriptome Profiling Analysis of Wolf Spider Pardosa pseudoannulata (Araneae: Lycosidae) after Cadmium Exposure. Int J Mol Sci 2016; 17:ijms17122033. [PMID: 27918488 PMCID: PMC5187833 DOI: 10.3390/ijms17122033] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 11/23/2016] [Accepted: 11/29/2016] [Indexed: 12/18/2022] Open
Abstract
Pardosa pseudoannulata is one of the most common wandering spiders in agricultural fields and a potentially good bioindicator for heavy metal contamination. However, little is known about the mechanisms by which spiders respond to heavy metals at the molecular level. In the present study, high-throughput transcriptome sequencing was employed to characterize the de novo transcriptome of the spiders and to identify differentially expressed genes (DEGs) after cadmium exposure. We obtained 60,489 assembled unigenes, 18,773 of which were annotated in the public databases. A total of 2939 and 2491 DEGs were detected between the libraries of two Cd-treated groups and the control. Functional enrichment analysis revealed that metabolism processes and digestive system function were predominately enriched in response to Cd stress. At the cellular and molecular levels, significantly enriched pathways in lysosomes and phagosomes as well as replication, recombination and repair demonstrated that oxidative damage resulted from Cd exposure. Based on the selected DEGs, certain critical genes involved in defence and detoxification were analysed. These results may elucidate the molecular mechanisms underlying spiders' responses to heavy metal stress.
Collapse
|
38
|
Khlifi R, Olmedo P, Gil F, Hammami B, Hamza-Chaffai A, Rebai A. Gene-environment interactions between ERCC2, ERCC3, XRCC1 and cadmium exposure in nasal polyposis disease. J Appl Genet 2016; 58:221-229. [PMID: 27838878 DOI: 10.1007/s13353-016-0375-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 11/01/2016] [Accepted: 11/03/2016] [Indexed: 11/27/2022]
Abstract
Gene-environment interactions have long been known to play an important role in complex disease aetiology, such as nasal polyposis (NP). The present study supports the concept that DNA repair gene polymorphisms play critical roles in modifying individual susceptibility to environmental diseases. In fact, we investigated the role of polymorphisms in DNA repair genes and cadmium as risk factors for Tunisian patients with NP. To the best of our knowledge, this is the first report on the impact of combined effects of cadmium and ERCC3 7122 A>G (rs4150407), ERCC2 Lys751Gln (rs13181) and XRCC1 Arg399Gln (rs25487) genes in the susceptibility to NP disease. Significant associations between the risk of developing NP disease and ERCC2 [odds ratio (OR) = 2.0, 95 % confidence interval (CI) = 1.1-3.7, p = 0.023] and ERCC3 (OR = 2.2, 95 % CI = 1.2-4.1, p = 0.013) genotypes polymorphisms were observed. Blood concentrations of Cd in NP patients (2.2 μg/L) were significantly higher than those of controls (0.5 μg/L). A significant interaction between ERCC3 (7122 A>G) polymorphism and blood-Cd levels (for the median of blood-Cd levels: OR = 3.8, 95 % CI = 1.3-10.8, p = 0.014 and for the 75th percentiles of blood-Cd levels: OR = 2.7, 95 % CI = 1.1-7.2, p = 0.041) was found in association with the risk of NP disease. In addition, when we stratified ERCC2, ERCC3 and XRCC1 polymorphism genotypes by the median and 75th percentiles of blood-Cd levels, we found also significant interactions between ERCC2 (Lys751Gln) and ERCC3 (7122 A>G) genotypes polymorphism and this metal in association with NP disease. However, no interaction was found between XRCC1 (Arg399Gln) polymorphism genotypes and Cd in association with NP disease.
Collapse
Affiliation(s)
- Rim Khlifi
- Unit of Marine and Environmental Toxicology, UR 09-03, Institut Préparatoire aux Etudes d'Ingénieur de Sfax (IPEIS), Sfax, Tunisia.
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, Sfax, Tunisia.
| | - Pablo Olmedo
- Department of Legal Medicine and Toxicology, University of Granada, Granada, Spain
| | - Fernando Gil
- Department of Legal Medicine and Toxicology, University of Granada, Granada, Spain
| | - Boutheina Hammami
- Department of Otorhinolaryngology, Habib Bourguiba Hospital, Sfax, Tunisia
| | - Amel Hamza-Chaffai
- Unit of Marine and Environmental Toxicology, UR 09-03, Institut Préparatoire aux Etudes d'Ingénieur de Sfax (IPEIS), Sfax, Tunisia
| | - Ahmed Rebai
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, Sfax, Tunisia
| |
Collapse
|
39
|
Kominkova M, Milosavljevic V, Vitek P, Polanska H, Cihalova K, Dostalova S, Hynstova V, Guran R, Kopel P, Richtera L, Masarik M, Brtnicky M, Kynicky J, Zitka O, Adam V. Comparative study on toxicity of extracellularly biosynthesized and laboratory synthesized CdTe quantum dots. J Biotechnol 2016; 241:193-200. [PMID: 27984119 DOI: 10.1016/j.jbiotec.2016.10.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 10/25/2016] [Accepted: 10/28/2016] [Indexed: 12/25/2022]
Abstract
Nanobiosynthesis belongs to the most recent methods for synthesis of nanoparticles. This type of synthesis provides many advantages including the uniformity in particle shape and size. The biosynthesis has also a significant advantage regarding chemical properties of the obtained particles. In this study, we characterized the basic properties and composition of quantum dots (QDs), obtained by the extracellular biosynthesis by Escherichia coli. Furthermore, the toxicity of the biosynthesized QDs was compared to QDs prepared by microwave synthesis. The obtained results revealed the presence of cyan CdTe QDs after removal of substantial amounts of organic compounds, which stabilized the nanoparticle surface. QDs toxicity was evaluated using three cell lines Human Foreskin Fibroblast (HFF), Human Prostate Cancer cells (PC-3) and Breast Cancer cells (MCF-7) and the MTT assay. The test revealed differences in the toxicity between variants of QDs, varying about 10% in the HFF and 30% in the MCF-7 cell lines. The toxicity of the biosynthesized QDs to the PC-3 cell lines was about 35% lower in comparison with the QDs prepared by microwave synthesis.
Collapse
Affiliation(s)
- Marketa Kominkova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic.
| | - Vedran Milosavljevic
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic.
| | - Petr Vitek
- Global Change Research Institute, The Czech Academy of Sciences, v.v.i., Belidla 4a, CZ-603 00 Brno, Czech Republic.
| | - Hana Polanska
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic; Department of Physiology and Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00 Brno, Czech Republic.
| | - Kristyna Cihalova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic.
| | - Simona Dostalova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic.
| | - Veronika Hynstova
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic.
| | - Roman Guran
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic.
| | - Pavel Kopel
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic.
| | - Lukas Richtera
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic.
| | - Michal Masarik
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic; Department of Physiology and Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-625 00 Brno, Czech Republic.
| | - Martin Brtnicky
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic; Department of Geology and Pedology, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic.
| | - Jindrich Kynicky
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic; Department of Geology and Pedology, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic.
| | - Ondrej Zitka
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic.
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00 Brno, Czech Republic.
| |
Collapse
|
40
|
Ototoxicity of Divalent Metals. Neurotox Res 2016; 30:268-82. [DOI: 10.1007/s12640-016-9627-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 04/21/2016] [Accepted: 04/22/2016] [Indexed: 12/16/2022]
|
41
|
Lee JY, Tokumoto M, Fujiwara Y, Hasegawa T, Seko Y, Shimada A, Satoh M. Accumulation of p53 via down-regulation of UBE2D family genes is a critical pathway for cadmium-induced renal toxicity. Sci Rep 2016; 6:21968. [PMID: 26912277 PMCID: PMC4766413 DOI: 10.1038/srep21968] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 02/03/2016] [Indexed: 12/19/2022] Open
Abstract
Chronic cadmium (Cd) exposure can induce renal toxicity. In Cd renal toxicity, p53 is thought to be involved. Our previous studies showed that Cd down-regulated gene expression of the UBE2D (ubiquitin-conjugating enzyme E2D) family members. Here, we aimed to define the association between UBE2D family members and p53-dependent apoptosis in human proximal tubular cells (HK-2 cells) treated with Cd. Cd increased intracellular p53 protein levels and decreased UBE2D2 and UBE2D4 gene expression via inhibition of YY1 and FOXF1 transcription factor activities. Double knockdown of UBE2D2 and UBE2D4 caused an increase in p53 protein levels, and knockdown of p53 attenuated not only Cd-induced apoptosis, but also Cd-induced apoptosis-related gene expression (BAX and PUMA). Additionally, the mice exposed to Cd for 6 months resulted in increased levels of p53 and induction of apoptosis in proximal tubular cells. These findings suggest that down-regulation of UBE2D family genes followed by accumulation of p53 in proximal tubular cells is an important mechanism for Cd-induced renal toxicity.
Collapse
Affiliation(s)
- Jin-Yong Lee
- Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan
| | - Maki Tokumoto
- Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan
| | - Yasuyuki Fujiwara
- Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan.,Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Tatsuya Hasegawa
- Department of Environmental Biochemistry, Mount Fuji Research Institute, 5597-1 Kenmarubi, Kamiyoshida, Fujiyoshida, Yamanashi 403-0005, Japan
| | - Yoshiyuki Seko
- Department of Environmental Biochemistry, Mount Fuji Research Institute, 5597-1 Kenmarubi, Kamiyoshida, Fujiyoshida, Yamanashi 403-0005, Japan
| | - Akinori Shimada
- Laboratory of Pathology, Department of Medical Technology, School of Life and Environmental Science, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa 252-5201, Japan
| | - Masahiko Satoh
- Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan
| |
Collapse
|
42
|
Pusnik M, Imeri M, Deppierraz G, Bruinink A, Zinn M. The agar diffusion scratch assay--A novel method to assess the bioactive and cytotoxic potential of new materials and compounds. Sci Rep 2016; 6:20854. [PMID: 26861591 PMCID: PMC4748257 DOI: 10.1038/srep20854] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 01/07/2016] [Indexed: 12/16/2022] Open
Abstract
A profound in vitro evaluation not only of the cytotoxic but also of bioactive potential of a given compound or material is crucial for predicting potential effects in the in vivo situation. However, most of the current methods have weaknesses in either the quantitative or qualitative assessment of cytotoxicity and/or bioactivity of the test compound. Here we describe a novel assay combining the ISO 10993-5 agar diffusion test and the scratch also termed wound healing assay. In contrast to these original tests this assay is able to detect and distinguish between cytotoxic, cell migration modifying and cytotoxic plus cell migration modifying compounds, and this at higher sensitivity and in a quantitative way.
Collapse
Affiliation(s)
- Mascha Pusnik
- Institute of Life Technologies, HES-SO Valais-Wallis, CH-1950 Sion, Switzerland
| | - Minire Imeri
- Institute of Life Technologies, HES-SO Valais-Wallis, CH-1950 Sion, Switzerland
| | - Grégoire Deppierraz
- Institute of Life Technologies, HES-SO Valais-Wallis, CH-1950 Sion, Switzerland
| | - Arie Bruinink
- Laboratory for Biointerfaces, Swiss Federal Laboratories for Materials Science and Technology - Empa, CH-9014 St. Gallen, Switzerland
| | - Manfred Zinn
- Institute of Life Technologies, HES-SO Valais-Wallis, CH-1950 Sion, Switzerland
| |
Collapse
|
43
|
Goodson WH, Lowe L, Carpenter DO, Gilbertson M, Manaf Ali A, Lopez de Cerain Salsamendi A, Lasfar A, Carnero A, Azqueta A, Amedei A, Charles AK, Collins AR, Ward A, Salzberg AC, Colacci A, Olsen AK, Berg A, Barclay BJ, Zhou BP, Blanco-Aparicio C, Baglole CJ, Dong C, Mondello C, Hsu CW, Naus CC, Yedjou C, Curran CS, Laird DW, Koch DC, Carlin DJ, Felsher DW, Roy D, Brown DG, Ratovitski E, Ryan EP, Corsini E, Rojas E, Moon EY, Laconi E, Marongiu F, Al-Mulla F, Chiaradonna F, Darroudi F, Martin FL, Van Schooten FJ, Goldberg GS, Wagemaker G, Nangami GN, Calaf GM, Williams G, Wolf GT, Koppen G, Brunborg G, Lyerly HK, Krishnan H, Ab Hamid H, Yasaei H, Sone H, Kondoh H, Salem HK, Hsu HY, Park HH, Koturbash I, Miousse IR, Scovassi AI, Klaunig JE, Vondráček J, Raju J, Roman J, Wise JP, Whitfield JR, Woodrick J, Christopher JA, Ochieng J, Martinez-Leal JF, Weisz J, Kravchenko J, Sun J, Prudhomme KR, Narayanan KB, Cohen-Solal KA, Moorwood K, Gonzalez L, Soucek L, Jian L, D'Abronzo LS, Lin LT, Li L, Gulliver L, McCawley LJ, Memeo L, Vermeulen L, Leyns L, Zhang L, Valverde M, Khatami M, Romano MF, Chapellier M, Williams MA, Wade M, Manjili MH, Lleonart ME, Xia M, Gonzalez MJ, Karamouzis MV, Kirsch-Volders M, Vaccari M, Kuemmerle NB, Singh N, Cruickshanks N, Kleinstreuer N, van Larebeke N, Ahmed N, Ogunkua O, Krishnakumar PK, Vadgama P, Marignani PA, Ghosh PM, Ostrosky-Wegman P, Thompson PA, Dent P, Heneberg P, Darbre P, Sing Leung P, Nangia-Makker P, Cheng QS, Robey RB, Al-Temaimi R, Roy R, Andrade-Vieira R, Sinha RK, Mehta R, Vento R, Di Fiore R, Ponce-Cusi R, Dornetshuber-Fleiss R, Nahta R, Castellino RC, Palorini R, Abd Hamid R, Langie SAS, Eltom SE, Brooks SA, Ryeom S, Wise SS, Bay SN, Harris SA, Papagerakis S, Romano S, Pavanello S, Eriksson S, Forte S, Casey SC, Luanpitpong S, Lee TJ, Otsuki T, Chen T, Massfelder T, Sanderson T, Guarnieri T, Hultman T, Dormoy V, Odero-Marah V, Sabbisetti V, Maguer-Satta V, Rathmell WK, Engström W, Decker WK, Bisson WH, Rojanasakul Y, Luqmani Y, Chen Z, Hu Z. Assessing the carcinogenic potential of low-dose exposures to chemical mixtures in the environment: the challenge ahead. Carcinogenesis 2015; 36 Suppl 1:S254-96. [PMID: 26106142 PMCID: PMC4480130 DOI: 10.1093/carcin/bgv039] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Low-dose exposures to common environmental chemicals that are deemed safe individually may be combining to instigate carcinogenesis, thereby contributing to the incidence of cancer. This risk may be overlooked by current regulatory practices and needs to be vigorously investigated. Lifestyle factors are responsible for a considerable portion of cancer incidence worldwide, but credible estimates from the World Health Organization and the International Agency for Research on Cancer (IARC) suggest that the fraction of cancers attributable to toxic environmental exposures is between 7% and 19%. To explore the hypothesis that low-dose exposures to mixtures of chemicals in the environment may be combining to contribute to environmental carcinogenesis, we reviewed 11 hallmark phenotypes of cancer, multiple priority target sites for disruption in each area and prototypical chemical disruptors for all targets, this included dose-response characterizations, evidence of low-dose effects and cross-hallmark effects for all targets and chemicals. In total, 85 examples of chemicals were reviewed for actions on key pathways/mechanisms related to carcinogenesis. Only 15% (13/85) were found to have evidence of a dose-response threshold, whereas 59% (50/85) exerted low-dose effects. No dose-response information was found for the remaining 26% (22/85). Our analysis suggests that the cumulative effects of individual (non-carcinogenic) chemicals acting on different pathways, and a variety of related systems, organs, tissues and cells could plausibly conspire to produce carcinogenic synergies. Additional basic research on carcinogenesis and research focused on low-dose effects of chemical mixtures needs to be rigorously pursued before the merits of this hypothesis can be further advanced. However, the structure of the World Health Organization International Programme on Chemical Safety ‘Mode of Action’ framework should be revisited as it has inherent weaknesses that are not fully aligned with our current understanding of cancer biology.
Collapse
Affiliation(s)
- William H Goodson
- California Pacific Medical Center Research Institute, 2100 Webster Street #401, San Francisco, CA 94115, USA, Getting to Know Cancer, Room 229A, 36 Arthur Street, Truro, Nova Scotia B2N 1X5, Canada, Lancaster Environment Centre, Lancaster University, Bailrigg, Lancaster LA1 4AP, UK, Institute for Health and the Environment, University at Albany, 5 University Pl., Rensselaer, NY 12144, USA, Getting to Know Cancer, Guelph N1G 1E4, Canada, School of Biotechnology, Faculty of Agriculture Biotechnology and Food Sciences, Sultan Zainal Abidin University, Tembila Campus, 22200 Besut, Terengganu, Malaysia, Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Navarra, Pamplona 31008, Spain, Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers, State University of New Jersey, Piscataway, NJ 08854, USA, Instituto de Biomedicina de Sevilla, Consejo Superior de Investigaciones Cientificas. Hospital Universitario Virgen del Rocio, Univ. de Sevilla., Avda Manuel Siurot sn. 41013 Sevilla, Spain, Department of Experimental and Clinical Medicine, University of Firenze, Florence 50134, Italy, School of Biological Sciences, University of Reading, Hopkins Building, Reading, Berkshire RG6 6UB, UK, Department of Nutrition, University of Oslo, Oslo, Norway, Department of Biochemistry and Biology, University of Bath, Claverton Down, Bath BA2 7AY, UK, Department of Public Health Sciences, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy, Department of Chemicals and Radiation, Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo N-0403, Norway, Planet Biotechnologies Inc., St Albert, Alberta T8N 5K4, Canada, Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40508, USA, Spanish National Cancer Research Centre, CNI
| | - Leroy Lowe
- Getting to Know Cancer, Room 229A, 36 Arthur Street, Truro, Nova Scotia B2N 1X5, Canada, Lancaster Environment Centre, Lancaster University, Bailrigg, Lancaster LA1 4AP, UK
| | - David O Carpenter
- Institute for Health and the Environment, University at Albany, 5 University Pl., Rensselaer, NY 12144, USA
| | | | - Abdul Manaf Ali
- School of Biotechnology, Faculty of Agriculture Biotechnology and Food Sciences, Sultan Zainal Abidin University, Tembila Campus, 22200 Besut, Terengganu, Malaysia
| | | | - Ahmed Lasfar
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers, State University of New Jersey, Piscataway, NJ 08854, USA
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, Consejo Superior de Investigaciones Cientificas. Hospital Universitario Virgen del Rocio, Univ. de Sevilla., Avda Manuel Siurot sn. 41013 Sevilla, Spain
| | - Amaya Azqueta
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Navarra, Pamplona 31008, Spain
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, Florence 50134, Italy
| | - Amelia K Charles
- School of Biological Sciences, University of Reading, Hopkins Building, Reading, Berkshire RG6 6UB, UK
| | | | - Andrew Ward
- Department of Biochemistry and Biology, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Anna C Salzberg
- Department of Public Health Sciences, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| | - Annamaria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | - Ann-Karin Olsen
- Department of Chemicals and Radiation, Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo N-0403, Norway
| | - Arthur Berg
- Department of Public Health Sciences, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| | - Barry J Barclay
- Planet Biotechnologies Inc., St Albert, Alberta T8N 5K4, Canada
| | - Binhua P Zhou
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40508, USA
| | - Carmen Blanco-Aparicio
- Spanish National Cancer Research Centre, CNIO, Melchor Fernandez Almagro, 3, 28029 Madrid, Spain
| | - Carolyn J Baglole
- Department of Medicine, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - Chenfang Dong
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40508, USA
| | - Chiara Mondello
- Istituto di Genetica Molecolare, CNR, Via Abbiategrasso 207, 27100 Pavia, Italy
| | - Chia-Wen Hsu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892-3375, USA
| | - Christian C Naus
- Department of Cellular and Physiological Sciences, Life Sciences Institute, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| | - Clement Yedjou
- Department of Biology, Jackson State University, Jackson, MS 39217, USA
| | - Colleen S Curran
- Department of Molecular and Environmental Toxicology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dale W Laird
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario N6A 3K7, Canada
| | - Daniel C Koch
- Stanford University Department of Medicine, Division of Oncology, Stanford, CA 94305, USA
| | - Danielle J Carlin
- Superfund Research Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27560, USA
| | - Dean W Felsher
- Department of Medicine, Oncology and Pathology, Stanford University, Stanford, CA 94305, USA
| | - Debasish Roy
- Department of Natural Science, The City University of New York at Hostos Campus, Bronx, NY 10451, USA
| | - Dustin G Brown
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523-1680, USA
| | - Edward Ratovitski
- Department of Head and Neck Surgery/Head and Neck Cancer Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523-1680, USA
| | - Emanuela Corsini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Emilio Rojas
- Department of Genomic Medicine and Environmental Toxicology, Institute for Biomedical Research, National Autonomous University of Mexico, Mexico City 04510, México
| | - Eun-Yi Moon
- Department of Bioscience and Biotechnology, Sejong University, Seoul 143-747, Korea
| | - Ezio Laconi
- Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Fabio Marongiu
- Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | - Ferdinando Chiaradonna
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy, SYSBIO Centre of Systems Biology, Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | - Firouz Darroudi
- Human Safety and Environmental Research, Department of Health Sciences, College of North Atlantic, Doha 24449, State of Qatar
| | - Francis L Martin
- Lancaster Environment Centre, Lancaster University, Bailrigg, Lancaster LA1 4AP, UK
| | - Frederik J Van Schooten
- Department of Toxicology, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University, Maastricht 6200, The Netherlands
| | - Gary S Goldberg
- Department of Molecular Biology, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA
| | - Gerard Wagemaker
- Hacettepe University, Center for Stem Cell Research and Development, Ankara 06640, Turkey
| | - Gladys N Nangami
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
| | - Gloria M Calaf
- Center for Radiological Research, Columbia University Medical Center, New York, NY 10032, USA, Instituto de Alta Investigacion, Universidad de Tarapaca, Arica, Chile
| | - Graeme Williams
- School of Biological Sciences, University of Reading, Reading, RG6 6UB, UK
| | - Gregory T Wolf
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Gudrun Koppen
- Environmental Risk and Health Unit, Flemish Institute for Technological Research, 2400 Mol, Belgium
| | - Gunnar Brunborg
- Department of Chemicals and Radiation, Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo N-0403, Norway
| | - H Kim Lyerly
- Department of Surgery, Pathology, Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Harini Krishnan
- Department of Molecular Biology, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA
| | - Hasiah Ab Hamid
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, 43400 Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Hemad Yasaei
- Department of Life Sciences, College of Health and Life Sciences and the Health and Environment Theme, Institute of Environment, Health and Societies, Brunel University Kingston Lane, Uxbridge, Middlesex UB8 3PH, UK
| | - Hideko Sone
- National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, Ibraki 3058506, Japan
| | - Hiroshi Kondoh
- Department of Geriatric Medicine, Kyoto University Hospital 54 Kawaharacho, Shogoin, Sakyo-ku Kyoto, 606-8507, Japan
| | - Hosni K Salem
- Department of Urology, Kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 11559, Egypt
| | - Hsue-Yin Hsu
- Department of Life Sciences, Tzu-Chi University, Hualien 970, Taiwan
| | - Hyun Ho Park
- School of Biotechnology, Yeungnam University, Gyeongbuk 712-749, South Korea
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Isabelle R Miousse
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - A Ivana Scovassi
- Istituto di Genetica Molecolare, CNR, Via Abbiategrasso 207, 27100 Pavia, Italy
| | - James E Klaunig
- Department of Environmental Health, Indiana University, School of Public Health, Bloomington, IN 47405, USA
| | - Jan Vondráček
- Department of Cytokinetics, Institute of Biophysics Academy of Sciences of the Czech Republic, Brno, CZ-61265, Czech Republic
| | - Jayadev Raju
- Regulatory Toxicology Research Division, Bureau of Chemical Safety, Food Directorate, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Jesse Roman
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA, Robley Rex VA Medical Center, Louisville, KY 40202, USA
| | - John Pierce Wise
- Department of Applied Medical Sciences, University of Southern Maine, 96 Falmouth St., Portland, ME 04104, USA
| | - Jonathan R Whitfield
- Mouse Models of Cancer Therapies Group, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Jordan Woodrick
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - Joseph A Christopher
- Cancer Research UK. Cambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UK
| | - Josiah Ochieng
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
| | | | - Judith Weisz
- Departments of Obstetrics and Gynecology and Pathology, Pennsylvania State University College of Medicine, Hershey PA 17033, USA
| | - Julia Kravchenko
- Department of Surgery, Pathology, Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jun Sun
- Department of Biochemistry, Rush University, Chicago, IL 60612, USA
| | - Kalan R Prudhomme
- Environmental and Molecular Toxicology, Environmental Health Science Center, Oregon State University, Corvallis, OR 97331, USA
| | | | - Karine A Cohen-Solal
- Department of Medicine/Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
| | - Kim Moorwood
- Department of Biochemistry and Biology, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Laetitia Gonzalez
- Laboratory for Cell Genetics, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Laura Soucek
- Mouse Models of Cancer Therapies Group, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain, Catalan Institution for Research and Advanced Studies (ICREA), Barcelona 08010, Spain
| | - Le Jian
- School of Public Health, Curtin University, Bentley, WA 6102, Australia, Department of Urology, University of California Davis, Sacramento, CA 95817, USA
| | - Leandro S D'Abronzo
- Department of Urology, University of California Davis, Sacramento, CA 95817, USA
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Lin Li
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, The People's Republic of China
| | - Linda Gulliver
- Faculty of Medicine, University of Otago, Dunedin 9054, New Zealand
| | - Lisa J McCawley
- Department of Biomedical Engineering and Cancer Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - Lorenzo Memeo
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Via Penninazzo 7, Viagrande (CT) 95029, Italy
| | - Louis Vermeulen
- Center for Experimental Molecular Medicine, Academic Medical Center, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
| | - Luc Leyns
- Laboratory for Cell Genetics, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Luoping Zhang
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA 94720-7360, USA
| | - Mahara Valverde
- Department of Genomic Medicine and Environmental Toxicology, Institute for Biomedical Research, National Autonomous University of Mexico, Mexico City 04510, México
| | - Mahin Khatami
- Inflammation and Cancer Research, National Cancer Institute (NCI) (Retired), National Institutes of Health, Bethesda, MD 20892, USA
| | - Maria Fiammetta Romano
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, 80131 Naples, Italy
| | - Marion Chapellier
- Centre De Recherche En Cancerologie, De Lyon, Lyon, U1052-UMR5286, France
| | - Marc A Williams
- United States Army Institute of Public Health, Toxicology Portfolio-Health Effects Research Program, Aberdeen Proving Ground, Edgewood, MD 21010-5403, USA
| | - Mark Wade
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia, Via Adamello 16, 20139 Milano, Italy
| | - Masoud H Manjili
- Department of Microbiology and Immunology, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA 23298, USA
| | - Matilde E Lleonart
- Institut De Recerca Hospital Vall D'Hebron, Passeig Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Menghang Xia
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892-3375, USA
| | - Michael J Gonzalez
- University of Puerto Rico, Medical Sciences Campus, School of Public Health, Nutrition Program, San Juan 00921, Puerto Rico
| | - Michalis V Karamouzis
- Department of Biological Chemistry, Medical School, University of Athens, Institute of Molecular Medicine and Biomedical Research, 10676 Athens, Greece
| | | | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | - Nancy B Kuemmerle
- Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Neetu Singh
- Advanced Molecular Science Research Centre (Centre for Advanced Research), King George's Medical University, Lucknow, Uttar Pradesh 226 003, India
| | - Nichola Cruickshanks
- Departments of Neurosurgery and Biochemistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Nicole Kleinstreuer
- Integrated Laboratory Systems Inc., in support of the National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods, RTP, NC 27709, USA
| | - Nik van Larebeke
- Analytische, Milieu en Geochemie, Vrije Universiteit Brussel, Brussel B1050, Belgium
| | - Nuzhat Ahmed
- Department of Obstetrics and Gynecology, University of Melbourne, Victoria 3052, Australia
| | - Olugbemiga Ogunkua
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
| | - P K Krishnakumar
- Center for Environment and Water, Research Institute, King Fahd University of Petroleum and Minerals, Dhahran 3126, Saudi Arabia
| | - Pankaj Vadgama
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK
| | - Paola A Marignani
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Paramita M Ghosh
- Department of Urology, University of California Davis, Sacramento, CA 95817, USA
| | - Patricia Ostrosky-Wegman
- Department of Genomic Medicine and Environmental Toxicology, Institute for Biomedical Research, National Autonomous University of Mexico, Mexico City 04510, México
| | - Patricia A Thompson
- Department of Pathology, Stony Brook School of Medicine, Stony Brook University, The State University of New York, Stony Brook, NY 11794-8691, USA
| | - Paul Dent
- Departments of Neurosurgery and Biochemistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Petr Heneberg
- Charles University in Prague, Third Faculty of Medicine, CZ-100 00 Prague 10, Czech Republic
| | - Philippa Darbre
- School of Biological Sciences, The University of Reading, Whiteknights, Reading RG6 6UB, England
| | - Po Sing Leung
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, The People's Republic of China
| | | | - Qiang Shawn Cheng
- Computer Science Department, Southern Illinois University, Carbondale, IL 62901, USA
| | - R Brooks Robey
- White River Junction Veterans Affairs Medical Center, White River Junction, VT 05009, USA, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Rabeah Al-Temaimi
- Human Genetics Unit, Department of Pathology, Faculty of Medicine, Kuwait University, Jabriya 13110, Kuwait
| | - Rabindra Roy
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - Rafaela Andrade-Vieira
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Ranjeet K Sinha
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Rekha Mehta
- Regulatory Toxicology Research Division, Bureau of Chemical Safety, Food Directorate, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Renza Vento
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies, Polyclinic Plexus, University of Palermo, Palermo 90127, Italy , Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
| | - Riccardo Di Fiore
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies, Polyclinic Plexus, University of Palermo, Palermo 90127, Italy
| | | | - Rita Dornetshuber-Fleiss
- Department of Pharmacology and Toxicology, University of Vienna, Vienna A-1090, Austria, Institute of Cancer Research, Department of Medicine, Medical University of Vienna, Wien 1090, Austria
| | - Rita Nahta
- Departments of Pharmacology and Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA 30322, USA
| | - Robert C Castellino
- Division of Hematology and Oncology, Department of Pediatrics, Children's Healthcare of Atlanta, GA 30322, USA, Department of Pediatrics, Emory University School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Roberta Palorini
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy, SYSBIO Centre of Systems Biology, Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | - Roslida Abd Hamid
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, 43400 Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Sabine A S Langie
- Environmental Risk and Health Unit, Flemish Institute for Technological Research, 2400 Mol, Belgium
| | - Sakina E Eltom
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
| | - Samira A Brooks
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Sandra Ryeom
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sandra S Wise
- Department of Applied Medical Sciences, University of Southern Maine, 96 Falmouth St., Portland, ME 04104, USA
| | - Sarah N Bay
- Program in Genetics and Molecular Biology, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA 30322, USA
| | - Shelley A Harris
- Population Health and Prevention, Research, Prevention and Cancer Control, Cancer Care Ontario, Toronto, Ontario, M5G 2L7, Canada, Departments of Epidemiology and Occupational and Environmental Health, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, M5T 3M7, Canada
| | - Silvana Papagerakis
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Simona Romano
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, 80131 Naples, Italy
| | - Sofia Pavanello
- Department of Cardiac, Thoracic and Vascular Sciences, Unit of Occupational Medicine, University of Padova, Padova 35128, Italy
| | - Staffan Eriksson
- Department of Anatomy, Physiology and Biochemistry, The Swedish University of Agricultural Sciences, PO Box 7011, VHC, Almas Allé 4, SE-756 51, Uppsala, Sweden
| | - Stefano Forte
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Via Penninazzo 7, Viagrande (CT) 95029, Italy
| | - Stephanie C Casey
- Stanford University Department of Medicine, Division of Oncology, Stanford, CA 94305, USA
| | - Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Tae-Jin Lee
- Department of Anatomy, College of Medicine, Yeungnam University, Daegu 705-717, South Korea
| | - Takemi Otsuki
- Department of Hygiene, Kawasaki Medical School, Matsushima Kurashiki, Okayama 701-0192, Japan
| | - Tao Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, AR 72079, USA
| | - Thierry Massfelder
- INSERM U1113, team 3 'Cell Signalling and Communication in Kidney and Prostate Cancer', University of Strasbourg, Faculté de Médecine, 67085 Strasbourg, France
| | - Thomas Sanderson
- INRS-Institut Armand-Frappier, 531 Boulevard des Prairies, Laval, QC H7V 1B7, Canada
| | - Tiziana Guarnieri
- Department of Biology, Geology and Environmental Sciences, Alma Mater Studiorum Università di Bologna, Via Francesco Selmi, 3, 40126 Bologna, Italy, Center for Applied Biomedical Research, S. Orsola-Malpighi University Hospital, Via Massarenti, 9, 40126 Bologna, Italy, National Institute of Biostructures and Biosystems, Viale Medaglie d' Oro, 305, 00136 Roma, Italy
| | - Tove Hultman
- Department of Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Swedish University of Agricultural Sciences, PO Box 7028, 75007 Uppsala, Sweden
| | - Valérian Dormoy
- INSERM U1113, team 3 'Cell Signalling and Communication in Kidney and Prostate Cancer', University of Strasbourg, Faculté de Médecine, 67085 Strasbourg, France, Department of Cell and Developmental Biology, University of California, Irvine, CA 92697, USA
| | - Valerie Odero-Marah
- Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
| | - Venkata Sabbisetti
- Harvard Medical School/Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Veronique Maguer-Satta
- United States Army Institute of Public Health, Toxicology Portfolio-Health Effects Research Program, Aberdeen Proving Ground, Edgewood, MD 21010-5403, USA
| | - W Kimryn Rathmell
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Wilhelm Engström
- Department of Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Swedish University of Agricultural Sciences, PO Box 7028, 75007 Uppsala, Sweden
| | | | - William H Bisson
- Environmental and Molecular Toxicology, Environmental Health Science Center, Oregon State University, Corvallis, OR 97331, USA
| | - Yon Rojanasakul
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, 26506, USA
| | - Yunus Luqmani
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kuwait University, PO Box 24923, Safat 13110, Kuwait and
| | - Zhenbang Chen
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
| | - Zhiwei Hu
- Department of Surgery, The Ohio State University College of Medicine, The James Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
44
|
Valko M, Jomova K, Rhodes CJ, Kuča K, Musílek K. Redox- and non-redox-metal-induced formation of free radicals and their role in human disease. Arch Toxicol 2015; 90:1-37. [DOI: 10.1007/s00204-015-1579-5] [Citation(s) in RCA: 535] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 08/11/2015] [Indexed: 02/07/2023]
|
45
|
Phatak VM, Muller PAJ. Metal toxicity and the p53 protein: an intimate relationship. Toxicol Res (Camb) 2015. [DOI: 10.1039/c4tx00117f] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The relationship between p53, ROS and transition metals.
Collapse
|
46
|
Biology of the cell cycle inhibitor p21CDKN1A: molecular mechanisms and relevance in chemical toxicology. Arch Toxicol 2014; 89:155-78. [DOI: 10.1007/s00204-014-1430-4] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 12/03/2014] [Indexed: 02/07/2023]
|
47
|
El-Atta HMA, El-Bakary AA, Attia AM, Lotfy A, Khater SS, Elsamanoudy AZ, Abdalla HA. DNA fragmentation, caspase 3 and prostate-specific antigen genes expression induced by arsenic, cadmium, and chromium on nontumorigenic human prostate cells. Biol Trace Elem Res 2014; 162:95-105. [PMID: 25227780 DOI: 10.1007/s12011-014-0100-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 08/05/2014] [Indexed: 10/24/2022]
Abstract
Prostate cancer is one of the most common cancers and the second cause of cancer-related deaths among men. Metals are recognized as chemical carcinogens where chronic exposures to such metals are implicated in the development of cancer, including prostate cancer. This in vitro study demonstrates the relative death sensitivity of prostatic (RWPE-1) cells to arsenic (As), cadmium (Cd), and chromium (Cr) as environmental pollutants through its apoptotic effects and the effect of these chemicals on prostate-specific antigen (PSA) gene expression as a marker for their carcinogecity. RWPE-1 cells were divided into three groups that were treated with As, Cd, and Cr in three replicates, at three different concentrations for each metal for 48 h. A control group consisted of untreated RWPE1 cells was used. Apoptosis was assessed using comet assay and caspase 3 gene expression; meanwhile, PSA gene expression was evaluated by semiqualitative real-time PCR (RT-PCR). One of the novel findings of this study is that arsenic and cadmium at low concentrations decreased apoptosis of RWPE-1 cells in a concentration-dependent manner while chromium induced significant concentration-dependent increase in apoptosis. Yet, at the highest concentrations, apoptosis was relatively more induced by all chemicals. Arsenic was the most chemical inhibiting apoptosis in RWPE-1 cells at low concentration. While at the moderate and highest concentrations, cadmium was the most inhibiting chemical of RWPE-1 cells' apoptosis. No distinct differences between treated and untreated cells for PSA gene expression were observed. It can be concluded that As and Cd, at low concentrations, can reduce apoptosis of prostatic cells in a concentration-dependent manner while chromium induced it; however, all metal salts used in this study did not induce PSA gene expression.
Collapse
Affiliation(s)
- Hend M Abo El-Atta
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | | | | | | | | | | | | |
Collapse
|
48
|
YANG HAIBO, SONG WEI, CHENG MEIDIE, FAN HAIFANG, GU XU, QIAO YING, LU XIN, YU RUIHE, CHEN LANYING. Deoxycholic acid inhibits the growth of BGC-823 gastric carcinoma cells via a p53-mediated pathway. Mol Med Rep 2014; 11:2749-54. [DOI: 10.3892/mmr.2014.3004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 11/03/2014] [Indexed: 11/06/2022] Open
|
49
|
Adámik M, Bažantová P, Navrátilová L, Polášková A, Pečinka P, Holaňová L, Tichý V, Brázdová M. Impact of cadmium, cobalt and nickel on sequence-specific DNA binding of p63 and p73 in vitro and in cells. Biochem Biophys Res Commun 2014; 456:29-34. [PMID: 25446071 DOI: 10.1016/j.bbrc.2014.11.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 11/11/2014] [Indexed: 11/30/2022]
Abstract
Site-specific DNA recognition and binding activity belong to common attributes of all three members of tumor suppressor p53 family proteins: p53, p63 and p73. It was previously shown that heavy metals can affect p53 conformation, sequence-specific binding and suppress p53 response to DNA damage. Here we report for the first time that cadmium, nickel and cobalt, which have already been shown to disturb various DNA repair mechanisms, can also influence p63 and p73 sequence-specific DNA binding activity and transactivation of p53 family target genes. Based on results of electrophoretic mobility shift assay and luciferase reporter assay, we conclude that cadmium inhibits sequence-specific binding of all three core domains to p53 consensus sequences and abolishes transactivation of several promoters (e.g. BAX and MDM2) by 50μM concentrations. In the presence of specific DNA, all p53 family core domains were partially protected against loss of DNA binding activity due to cadmium treatment. Effective cadmium concentration to abolish DNA-protein interactions was about two times higher for p63 and p73 proteins than for p53. Furthermore, we detected partial reversibility of cadmium inhibition for all p53 family members by EDTA. DTT was able to reverse cadmium inhibition only for p53 and p73. Nickel and cobalt abolished DNA-p53 interaction at sub-millimolar concentrations while inhibition of p63 and p73 DNA binding was observed at millimolar concentrations. In summary, cadmium strongly inhibits p53, p63 and p73 DNA binding in vitro and in cells in comparison to nickel and cobalt. The role of cadmium inhibition of p53 tumor suppressor family in carcinogenesis is discussed.
Collapse
Affiliation(s)
- Matej Adámik
- Institute of Biophysics, Academy of Science of the Czech Republic, v.v.i., Královopolská 135, 612 65 Brno, Czech Republic
| | - Pavla Bažantová
- Institute of Biophysics, Academy of Science of the Czech Republic, v.v.i., Královopolská 135, 612 65 Brno, Czech Republic; Department of Biology and Ecology, Faculty of Science, University of Ostrava, Chittussiho 10, 701 03 Ostrava, Czech Republic
| | - Lucie Navrátilová
- Institute of Biophysics, Academy of Science of the Czech Republic, v.v.i., Královopolská 135, 612 65 Brno, Czech Republic
| | - Alena Polášková
- Institute of Biophysics, Academy of Science of the Czech Republic, v.v.i., Královopolská 135, 612 65 Brno, Czech Republic
| | - Petr Pečinka
- Institute of Biophysics, Academy of Science of the Czech Republic, v.v.i., Královopolská 135, 612 65 Brno, Czech Republic; Department of Biology and Ecology, Faculty of Science, University of Ostrava, Chittussiho 10, 701 03 Ostrava, Czech Republic
| | - Lucie Holaňová
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Palackého 1/3, 61242 Brno, Czech Republic
| | - Vlastimil Tichý
- Institute of Biophysics, Academy of Science of the Czech Republic, v.v.i., Královopolská 135, 612 65 Brno, Czech Republic
| | - Marie Brázdová
- Institute of Biophysics, Academy of Science of the Czech Republic, v.v.i., Královopolská 135, 612 65 Brno, Czech Republic; Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Palackého 1/3, 61242 Brno, Czech Republic.
| |
Collapse
|
50
|
Luevano J, Damodaran C. A review of molecular events of cadmium-induced carcinogenesis. J Environ Pathol Toxicol Oncol 2014; 33:183-94. [PMID: 25272057 DOI: 10.1615/jenvironpatholtoxicoloncol.2014011075] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cadmium (Cd) is a toxic, heavy industrial metal that poses serious environmental health hazards to both humans and wildlife. Recently, Cd and Cd-containing compounds have been classified as known human carcinogens, and epidemiological data show causal associations with prostate, breast, and lung cancer. The molecular mechanisms involved in Cd-induced carcinogenesis are poorly understood and are only now beginning to be elucidated. The effects of chronic exposure to Cd have recently attracted great interest due to the development of malignancies in Cd-induced tumorigenesis in animals models. Briefly, various in vitro studies demonstrate that Cd can act as a mitogen, can stimulate cell proliferation and inhibit apoptosis and DNA repair, and can induce carcinogenesis in several mammalian tissues and organs. Thus, the various mechanisms involved in chronic Cd exposure and malignant transformations warrant further investigation. In this review, we focus on recent evidence of various leading general and tissue-specific molecular mechanisms that follow chronic exposure to Cd in prostate-, breast-, and lung-transformed malignancies. In addition, in this review, we consider less defined mechanisms such as epigenetic modification and autophagy, which are thought to play a role in the development of Cd-induced malignant transformation.
Collapse
Affiliation(s)
- Joe Luevano
- Center of Excellence in Cancer Research, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | | |
Collapse
|