1
|
Li S, Liu Y, Wu Y, Ren L, Lu Y, Yamaguchi S, Lu Q, Hu C, Li D, Jiang N. An Outlook on Platinum-Based Active Ingredients for Dermatologic and Skincare Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1303. [PMID: 39120408 PMCID: PMC11314049 DOI: 10.3390/nano14151303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024]
Abstract
Platinum-based materials exhibit a broad spectrum of biological activities, including antioxidant, anti-inflammatory, antimicrobial, and pro-collagen synthesis properties, making them particularly useful for various biomedical applications. This review summarizes the biological effects and therapeutic potential of platinum-based active ingredients in dermatological and skincare applications. We discuss their synthesis methods and their antioxidant, anti-inflammatory, antimicrobial, and collagen synthesis properties, which play essential roles in treating skin conditions including psoriasis and acne, as well as enhancing skin aesthetics in anti-aging products. Safety and sustainability concerns, including the need for green synthesis and comprehensive toxicological assessments to ensure safe topical applications, are also discussed. By providing an up-to-date overview of current research, we aim to highlight both the potential and the current challenges of platinum-based active ingredients in advancing dermatology and skincare solutions.
Collapse
Affiliation(s)
- Shining Li
- Key Laboratory of Advanced Materials and Devices for Post-Moore Chips, Ministry of Education, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Yizhou Liu
- Key Laboratory of Advanced Materials and Devices for Post-Moore Chips, Ministry of Education, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Ying Wu
- Key Laboratory of Advanced Materials and Devices for Post-Moore Chips, Ministry of Education, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Lu Ren
- Hua An Tang Biotech Group Co., Ltd., Guangzhou 511434, China
| | - Yongjie Lu
- Hua An Tang Biotech Group Co., Ltd., Guangzhou 511434, China
| | | | - Qipeng Lu
- Key Laboratory of Advanced Materials and Devices for Post-Moore Chips, Ministry of Education, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Chuangang Hu
- State Key Laboratory of Organic–Inorganic Composites, College of Chemical Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Dongcui Li
- Hua An Tang Biotech Group Co., Ltd., Guangzhou 511434, China
| | - Naisheng Jiang
- Key Laboratory of Advanced Materials and Devices for Post-Moore Chips, Ministry of Education, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| |
Collapse
|
2
|
Kang YT, Yang WJ, Huang HC, Tang SC, Ko JL. Exposure to nickel chloride induces epigenetic modification on detoxification enzyme glutathione S-transferase M2. ENVIRONMENTAL TOXICOLOGY 2024; 39:1729-1736. [PMID: 38050843 DOI: 10.1002/tox.24055] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/23/2023] [Accepted: 11/12/2023] [Indexed: 12/07/2023]
Abstract
Nickel (Ni) is a human carcinogen with genotoxic and epigenotoxic effects. Environmental and occupational exposure to Ni increases the risk of cancer and chronic inflammatory diseases. Our previous findings indicate that Ni alters gene expression through epigenetic regulation, specifically impacting E-cadherin and angiopoietin-like 4 (ANGPTL4), involved in epithelial-mesenchymal transition and migration. GST-M2, a member of the glutathione S-transferase (GST) enzyme family, plays a crucial role in cellular defense against oxidative damage and has been increasingly associated with cancer. GST-M2 overexpression inhibits lung cancer invasion and metastasis in vitro and in vivo. Hypermethylation of its promoter in cancer cells reduces gene expression, correlating with poor prognosis in non-small-cell lung cancer patients. The impact of Ni on GST-M2 remains unclear. We will investigate whether nickel exerts regulatory effects on GST-M2 through epigenetic modifications. Additionally, metformin, an antidiabetic drug, is being studied as a chemopreventive agent against nickel-induced damage. Our findings indicate that nickel chloride (NiCl2 ) exposure, both short-term and long-term, represses GST-M2 expression. However, the expression can be restored by demethylation agent 5-aza-2'-deoxycytidine and metformin. NiCl2 promotes hypermethylation of the GST-M2 promoter, as confirmed by methylation-specific PCR and bisulfite sequencing. Additionally, NiCl2 also influences histone acetylation, and metformin counteracts the suppressive effect of NiCl2 on histone H3 expression. Metformin reestablishes the binding of specificity protein 1 to the GST-M2 promoter, which is otherwise disrupted by NiCl2 . These findings elucidate the mechanism by which Ni reduces GST-M2 expression and transcriptional activity, potentially contributing to Ni-induced lung carcinogenesis.
Collapse
Affiliation(s)
- Yu-Ting Kang
- Institute of Medicine, Chung-Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Wan-Jung Yang
- Institute of Medicine, Chung-Shan Medical University, Taichung, Taiwan
- Department of Genomic Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Hsu Chih Huang
- Institute of Medicine, Chung-Shan Medical University, Taichung, Taiwan
- Division of Thoracic Surgery, Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Sheau-Chung Tang
- Department of Nursing, National Taichung University of Science and Technology, Taichung, Taiwan
| | - Jiunn-Liang Ko
- Institute of Medicine, Chung-Shan Medical University, Taichung, Taiwan
- Department of Medical Oncology and Chest Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
3
|
South AP, Laimer M, Gueye M, Sui JY, Eichenfield LF, Mellerio JE, Nyström A. Type VII Collagen Deficiency in the Oncogenesis of Cutaneous Squamous Cell Carcinoma in Dystrophic Epidermolysis Bullosa. J Invest Dermatol 2023; 143:2108-2119. [PMID: 37327859 DOI: 10.1016/j.jid.2023.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/11/2023] [Accepted: 05/17/2023] [Indexed: 06/18/2023]
Abstract
Dystrophic epidermolysis bullosa is a rare genetic skin disorder caused by COL7A1 sequence variations that result in type VII collagen deficits and cutaneous and extracutaneous manifestations. One serious complication of dystrophic epidermolysis bullosa is cutaneous squamous cell carcinoma, a leading driver of morbidity and mortality, especially among patients with recessive dystrophic epidermolysis bullosa. Type VII collagen deficits alter TGFβ signaling and evoke multiple other cutaneous squamous cell carcinoma progression-promoting activities within epidermal microenvironments. This review examines cutaneous squamous cell carcinoma pathophysiology in dystrophic epidermolysis bullosa with a focus on known oncogenesis pathways at play and explores the idea that therapeutic type VII collagen replacement may reduce cutaneous squamous cell carcinoma risk.
Collapse
Affiliation(s)
- Andrew P South
- Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| | - Martin Laimer
- Department of Dermatology and Allergology and EB House Austria, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | | | - Jennifer Y Sui
- Departments of Dermatology and Pediatrics, University of California San Diego School of Medicine, San Diego, California, USA; Division of Pediatric Dermatology, Rady Children's Hospital San Diego, San Diego, California, USA
| | - Lawrence F Eichenfield
- Departments of Dermatology and Pediatrics, University of California San Diego School of Medicine, San Diego, California, USA; Division of Pediatric Dermatology, Rady Children's Hospital San Diego, San Diego, California, USA
| | - Jemima E Mellerio
- St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Alexander Nyström
- Department of Dermatology, Medical Faculty, Medical Center, University of Freiburg, Freiburg, Germany; Freiburg Institute for Advanced Studies, Freiburg, Germany
| |
Collapse
|
4
|
Gaspar AD, Cuddapah S. Nickel-induced alterations to chromatin structure and function. Toxicol Appl Pharmacol 2022; 457:116317. [PMID: 36400264 PMCID: PMC9722551 DOI: 10.1016/j.taap.2022.116317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022]
Abstract
Nickel (Ni), a heavy metal is prevalent in the atmosphere due to both natural and anthropogenic activities. Ni is a carcinogen implicated in the development of lung and nasal cancers in humans. Furthermore, Ni exposure is associated with a number of chronic lung diseases in humans including asthma, chronic bronchitis, emphysema, pulmonary fibrosis, pulmonary edema and chronic obstructive pulmonary disease (COPD). While Ni compounds are weak mutagens, a number of studies have demonstrated the potential of Ni to alter the epigenome, suggesting epigenomic dysregulation as an important underlying cause for its pathogenicity. In the eukaryotic nucleus, the DNA is organized in a three-dimensional (3D) space through assembly of higher order chromatin structures. Such an organization is critically important for transcription and other biological activities. Accumulating evidence suggests that by negatively affecting various cellular regulatory processes, Ni could potentially affect chromatin organization. In this review, we discuss the role of Ni in altering the chromatin architecture, which potentially plays a major role in Ni pathogenicity.
Collapse
Affiliation(s)
- Adrian Domnic Gaspar
- Division of Environmental Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10010, USA
| | - Suresh Cuddapah
- Division of Environmental Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10010, USA.
| |
Collapse
|
5
|
Zhao L, Islam R, Wang Y, Zhang X, Liu LZ. Epigenetic Regulation in Chromium-, Nickel- and Cadmium-Induced Carcinogenesis. Cancers (Basel) 2022; 14:cancers14235768. [PMID: 36497250 PMCID: PMC9737485 DOI: 10.3390/cancers14235768] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022] Open
Abstract
Environmental and occupational exposure to heavy metals, such as hexavalent chromium, nickel, and cadmium, are major health concerns worldwide. Some heavy metals are well-documented human carcinogens. Multiple mechanisms, including DNA damage, dysregulated gene expression, and aberrant cancer-related signaling, have been shown to contribute to metal-induced carcinogenesis. However, the molecular mechanisms accounting for heavy metal-induced carcinogenesis and angiogenesis are still not fully understood. In recent years, an increasing number of studies have indicated that in addition to genotoxicity and genetic mutations, epigenetic mechanisms play critical roles in metal-induced cancers. Epigenetics refers to the reversible modification of genomes without changing DNA sequences; epigenetic modifications generally involve DNA methylation, histone modification, chromatin remodeling, and non-coding RNAs. Epigenetic regulation is essential for maintaining normal gene expression patterns; the disruption of epigenetic modifications may lead to altered cellular function and even malignant transformation. Therefore, aberrant epigenetic modifications are widely involved in metal-induced cancer formation, development, and angiogenesis. Notably, the role of epigenetic mechanisms in heavy metal-induced carcinogenesis and angiogenesis remains largely unknown, and further studies are urgently required. In this review, we highlight the current advances in understanding the roles of epigenetic mechanisms in heavy metal-induced carcinogenesis, cancer progression, and angiogenesis.
Collapse
|
6
|
Manić L, Wallace D, Onganer PU, Taalab YM, Farooqi AA, Antonijević B, Buha Djordjevic A. Epigenetic mechanisms in metal carcinogenesis. Toxicol Rep 2022; 9:778-787. [PMID: 36561948 PMCID: PMC9764177 DOI: 10.1016/j.toxrep.2022.03.037] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/16/2022] [Accepted: 03/26/2022] [Indexed: 12/25/2022] Open
Abstract
Many metals exhibit genotoxic and/or carcinogenic effects. These toxic metals can be found ubiquitously - in drinking water, food, air, general use products, in everyday and occupational settings. Exposure to such carcinogenic metals can result in serious health disorders, including cancer. Arsenic, cadmium, chromium, nickel, and their compounds have already been recognized as carcinogens by the International Agency for Research on Cancer. This review summarizes a wide range of epigenetic mechanisms contributing to carcinogenesis induced by these metals, primarily including, but not limited to, DNA methylation, miRNA regulation, and histone posttranslational modifications. The mechanisms are described and discussed both from a metal-centric and a mechanism-centric standpoint. The review takes a broad perspective, putting the mechanisms in the context of real-life exposure, and aims to assist in guiding future research, particularly with respect to the assessment and control of exposure to carcinogenic metals and novel therapy development.
Collapse
Affiliation(s)
- Luka Manić
- Department of Toxicology “Akademik Danilo Soldatović”, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - David Wallace
- School of Biomedical Science, Oklahoma State University Center for Health Sciences, Tulsa, United States
| | - Pinar Uysal Onganer
- Cancer Research Group, School of Life Sciences, University of Westminster, London, UK
| | - Yasmeen M. Taalab
- Institute of Forensic and Traffic Medicine, University of Heidelberg, Voßstraße 2, 69115 Heidelberg, Germany,Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Mansoura University, Dakahlia Governate 35516, Egypt
| | - Ammad Ahmad Farooqi
- Laboratory for Translational Oncology and Personalized Medicine, RLMC, Lahore, Pakistan
| | - Biljana Antonijević
- Department of Toxicology “Akademik Danilo Soldatović”, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Buha Djordjevic
- Department of Toxicology “Akademik Danilo Soldatović”, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia,Correspondence to: Department of Toxicology “Akademik Danilo Soldatović”, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia.
| |
Collapse
|
7
|
Madhu NR, Sarkar B, Slama P, Jha NK, Ghorai SK, Jana SK, Govindasamy K, Massanyi P, Lukac N, Kumar D, Kalita JC, Kesari KK, Roychoudhury S. Effect of Environmental Stressors, Xenobiotics, and Oxidative Stress on Male Reproductive and Sexual Health. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1391:33-58. [PMID: 36472815 DOI: 10.1007/978-3-031-12966-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This article examines the environmental factor-induced oxidative stress (OS) and their effects on male reproductive and sexual health. There are several factors that induce OS, i.e. radition, metal contamination, xenobiotic compounds, and cigarette smoke and lead to cause toxicity in the cells through metabolic or bioenergetic processes. These environmental factors may produce free radicals and enhance the reactive oxygen species (ROS). Free radicals are molecules that include oxygen and disbalance the amount of electrons that can create major chemical chains in the body and cause oxidation. Oxidative damage to cells may impair male fertility and lead to abnormal embryonic development. Moreover, it does not only cause a vast number of health issues such as ageing, cancer, atherosclerosis, insulin resistance, diabetes mellitus, cardiovascular diseases, ischemia-reperfusion injury, and neurodegenerative disorders but also decreases the motility of spermatozoa while increasing sperm DNA damage, impairing sperm mitochondrial membrane lipids and protein kinases. This chapter mainly focuses on the environmental stressors with further discussion on the mechanisms causing congenital impairments due to poor sexual health and transmitting altered signal transduction pathways in male gonadal tissues.
Collapse
Affiliation(s)
- Nithar Ranjan Madhu
- Department of Zoology, Acharya Prafulla Chandra College, New Barrackpore, Kolkata, West Bengal, India
| | - Bhanumati Sarkar
- Department of Botany, Acharya Prafulla Chandra College, New Barrackpore, Kolkata, West Bengal, India
| | - Petr Slama
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, India
| | | | - Sandip Kumar Jana
- Department of Zoology, Bajkul Milani Mahavidyalaya, Purba Medinipur, West Bengal, India
| | - Kadirvel Govindasamy
- Animal Production Division, ICAR Research Complex for NEH Region, Indian Council of Agricultural Research, Umiam, Meghalaya, India
| | - Peter Massanyi
- Department of Animal Physiology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Nitra, Slovak Republic
| | - Norbert Lukac
- Department of Animal Physiology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Nitra, Slovak Republic
| | - Dhruv Kumar
- School of Health Sciences & Technology, UPES University, Dehradun, Uttarakhand, India
| | - Jogen C Kalita
- Department of Zoology, Gauhati University, Guwahati, India
| | | | | |
Collapse
|
8
|
Lee HW, Jose CC, Cuddapah S. Epithelial-mesenchymal transition: Insights into nickel-induced lung diseases. Semin Cancer Biol 2021; 76:99-109. [PMID: 34058338 PMCID: PMC8627926 DOI: 10.1016/j.semcancer.2021.05.020] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023]
Abstract
Nickel compounds are environmental toxicants, prevalent in the atmosphere due to their widespread use in several industrial processes, extensive consumption of nickel containing products, as well as burning of fossil fuels. Exposure to nickel is associated with a multitude of chronic inflammatory lung diseases including asthma, chronic obstructive pulmonary disease (COPD) and pulmonary fibrosis. In addition, nickel exposure is implicated in the development of nasal and lung cancers. Interestingly, a common pathogenic mechanism underlying the development of diseases associated with nickel exposure is epithelial-mesenchymal transition (EMT). EMT is a process by which the epithelial cells lose their junctions and polarity and acquire mesenchymal traits, including increased ability to migrate and invade. EMT is a normal and essential physiological process involved in differentiation, development and wound healing. However, EMT also contributes to a number of pathological conditions, including fibrosis, cancer and metastasis. Growing evidence suggest that EMT induction could be an important outcome of nickel exposure. In this review, we discuss the role of EMT in nickel-induced lung diseases and the mechanisms associated with EMT induction by nickel exposure.
Collapse
Affiliation(s)
- Hyun-Wook Lee
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, 10010, USA
| | - Cynthia C Jose
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, 10010, USA
| | - Suresh Cuddapah
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, 10010, USA.
| |
Collapse
|
9
|
EFSA Panel on Contaminants in the Food Chain (CONTAM), Schrenk D, Bignami M, Bodin L, Chipman JK, del Mazo J, Grasl‐Kraupp B, Hogstrand C, Hoogenboom L(R, Leblanc J, Nebbia CS, Ntzani E, Petersen A, Sand S, Schwerdtle T, Vleminckx C, Wallace H, Guérin T, Massanyi P, Van Loveren H, Baert K, Gergelova P, Nielsen E. Update of the risk assessment of nickel in food and drinking water. EFSA J 2020; 18:e06268. [PMID: 33193868 PMCID: PMC7643711 DOI: 10.2903/j.efsa.2020.6268] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The European Commission asked EFSA to update its previous Opinion on nickel in food and drinking water, taking into account new occurrence data, the updated benchmark dose (BMD) Guidance and newly available scientific information. More than 47,000 analytical results on the occurrence of nickel were used for calculating chronic and acute dietary exposure. An increased incidence of post-implantation loss in rats was identified as the critical effect for the risk characterisation of chronic oral exposure and a BMDL 10 of 1.3 mg Ni/kg body weight (bw) per day was selected as the reference point for the establishment of a tolerable daily intake (TDI) of 13 μg/kg bw. Eczematous flare-up reactions in the skin elicited in nickel-sensitised humans, a condition known as systemic contact dermatitis, was identified as the critical effect for the risk characterisation of acute oral exposure. A BMDL could not be derived, and therefore, the lowest-observed-adverse-effect-level of 4.3 μg Ni/kg bw was selected as the reference point. The margin of exposure (MOE) approach was applied and an MOE of 30 or higher was considered as being indicative of a low health concern. The mean lower bound (LB)/upper bound (UB) chronic dietary exposure was below or at the level of the TDI. The 95th percentile LB/UB chronic dietary exposure was below the TDI in adolescents and in all adult age groups, but generally exceeded the TDI in toddlers and in other children, as well as in infants in some surveys. This may raise a health concern in these young age groups. The MOE values for the mean UB acute dietary exposure and for the 95th percentile UB raises a health concern for nickel-sensitised individuals. The MOE values for an acute scenario regarding consumption of a glass of water on an empty stomach do not raise a health concern.
Collapse
|
10
|
Premi S. Role of Melanin Chemiexcitation in Melanoma Progression and Drug Resistance. Front Oncol 2020; 10:1305. [PMID: 32850409 PMCID: PMC7425655 DOI: 10.3389/fonc.2020.01305] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 06/23/2020] [Indexed: 01/26/2023] Open
Abstract
Melanoma is the deadliest type of skin cancer. Human melanomas often show hyperactivity of nitric oxide synthase (NOS) and NADPH oxidase (NOX), which, respectively, generate nitric oxide (NO · ) and superoxide (O2 ·- ). The NO · and O2 - react instantly with each other to generate peroxynitrite (ONOO-) which is the driver of melanin chemiexcitation. Melanoma precursors, the melanocytes, are specialized skin cells that synthesize melanin, a potent shield against sunlight's ultraviolet (UV) radiation. However, melanin chemiexcitation paradoxically demonstrates the melanomagenic properties of melanin. In a loop, the NOS activity regulates melanin synthesis, and melanin is utilized by the chemiexcitation pathway to generate carcinogenic melanin-carbonyls in an excited triplet state. These carbonyl compounds induce UV-specific DNA damage without UV. Additionally, the carbonyl compounds are highly reactive and can make melanomagenic adducts with proteins, DNA and other biomolecules. Here we review the role of the melanin chemiexcitation pathway in melanoma initiation, progression, and drug resistance. We conclude by hypothesizing a non-classical, positive loop in melanoma where melanin chemiexcitation generates carcinogenic reactive carbonyl species (RCS) and DNA damage in normal melanocytes. In parallel, NOS and NOX regulate melanin synthesis generating raw material for chemiexcitation, and the resulting RCS and reactive nitrogen species (RNS) regulate cellular proteome and transcriptome in favor of melanoma progression, metastasis, and resistance against targeted therapies.
Collapse
Affiliation(s)
- Sanjay Premi
- Department of Tumor Biology, Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| |
Collapse
|
11
|
Genchi G, Carocci A, Lauria G, Sinicropi MS, Catalano A. Nickel: Human Health and Environmental Toxicology. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E679. [PMID: 31973020 PMCID: PMC7037090 DOI: 10.3390/ijerph17030679] [Citation(s) in RCA: 570] [Impact Index Per Article: 114.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/14/2020] [Accepted: 01/18/2020] [Indexed: 12/20/2022]
Abstract
Nickel is a transition element extensively distributed in the environment, air, water, and soil. It may derive from natural sources and anthropogenic activity. Although nickel is ubiquitous in the environment, its functional role as a trace element for animals and human beings has not been yet recognized. Environmental pollution from nickel may be due to industry, the use of liquid and solid fuels, as well as municipal and industrial waste. Nickel contact can cause a variety of side effects on human health, such as allergy, cardiovascular and kidney diseases, lung fibrosis, lung and nasal cancer. Although the molecular mechanisms of nickel-induced toxicity are not yet clear, mitochondrial dysfunctions and oxidative stress are thought to have a primary and crucial role in the toxicity of this metal. Recently, researchers, trying to characterize the capability of nickel to induce cancer, have found out that epigenetic alterations induced by nickel exposure can perturb the genome. The purpose of this review is to describe the chemical features of nickel in human beings and the mechanisms of its toxicity. Furthermore, the attention is focused on strategies to remove nickel from the environment, such as phytoremediation and phytomining.
Collapse
Affiliation(s)
- Giuseppe Genchi
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione, Università della Calabria, 87036 Arcavacata di Rende (Cosenza), Italy; (G.G.); (G.L.)
| | - Alessia Carocci
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari “A. Moro”, 70125 Bari, Italy;
| | - Graziantonio Lauria
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione, Università della Calabria, 87036 Arcavacata di Rende (Cosenza), Italy; (G.G.); (G.L.)
| | - Maria Stefania Sinicropi
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione, Università della Calabria, 87036 Arcavacata di Rende (Cosenza), Italy; (G.G.); (G.L.)
| | - Alessia Catalano
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari “A. Moro”, 70125 Bari, Italy;
| |
Collapse
|
12
|
Son YO. Molecular Mechanisms of Nickel-Induced Carcinogenesis. Endocr Metab Immune Disord Drug Targets 2019; 20:1015-1023. [PMID: 31774048 DOI: 10.2174/1871530319666191125112728] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/11/2019] [Accepted: 03/22/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND The increased use of heavy metal nickel in modern industries results in increased environmental impact. Occupational and environmental exposure to nickel is closely linked to an increased risk of human lung cancer and nasal cancer. OBJECTIVE Unlike other heavy metal carcinogens, nickel has weak mutagenic activity. Carcinogenesis caused by nickel is intensively studied, but the precise mechanism of action is not yet known. RESULTS Epigenetic changes, activation of hypoxia signaling pathways, and generation of reactive oxygen species (ROS) are considered to be the major molecular mechanisms involved in nickelinduced carcinogenesis. CONCLUSION This review provides insights into current research on nickel-induced carcinogenesis and suggests possible effective therapeutic strategies for nickel-induced carcinogenesis.
Collapse
Affiliation(s)
- Young-Ok Son
- Department of Animal Biotechnology, Faculty of Biotechnology and Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju City, Jeju Special Self-Governing Province, 63243, Korea
| |
Collapse
|
13
|
Sun CQ, Arnold RS, Hsieh CL, Dorin JR, Lian F, Li Z, Petros JA. Discovery and mechanisms of host defense to oncogenesis: targeting the β-defensin-1 peptide as a natural tumor inhibitor. Cancer Biol Ther 2019; 20:774-786. [PMID: 30900935 PMCID: PMC6605992 DOI: 10.1080/15384047.2018.1564564] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 11/27/2018] [Accepted: 12/25/2018] [Indexed: 12/28/2022] Open
Abstract
Human beta-defensin-1 (hBD-1) is one of a number of small cationic host-defense peptides. Besides its well-known broad-spectrum antimicrobial function, hBD-1 has recently been identified as a chromosome 8p tumor-suppressor gene. The role of hBD-1 in modulating the host immune response to oncogenesis, associated with cell signaling and potential therapeutic applications, has become increasingly appreciated over time. In this study, multiple approaches were used to illustrate hBD-1 anti-tumor activities. Results demonstrate that hBD-1 peptide alters human epidermal growth factor receptor 2 (HER2) signal transduction and represses retroviral-mediated transgene expression in cancer cells. Loss of orthologous murine defense-1 (mBD1) in mice enhances nickel sulfate-induced leiomyosarcoma and causes mouse kidney cells to exhibit increased susceptibility to HPV-16 E6/7-induced neoplastic transformation. Furthermore, for the first time, a novel function of the urine-derived hBD-1 peptide was discovered to suppress bladder cancer growth and this may lead to future applications in the treatment of malignancy.
Collapse
Affiliation(s)
- Carrie Q. Sun
- Department of Urology and Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Rebecca S. Arnold
- Department of Urology and Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Chia-Ling Hsieh
- Department of Molecular Medicine, China Medical University Hospital, Taipei, Taiwan
| | - Julia R. Dorin
- Center for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Fei Lian
- Emory University School of Medicine, Emory University, Atlanta, Georgia
| | - Zhenghong Li
- School of Medicine, Central South University, Changsha City, Human Province, P. R. China
| | - John A. Petros
- Department of Urology and Winship Cancer Institute, Emory University, Atlanta, Georgia
- Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| |
Collapse
|
14
|
Role of Adiponectin in Endoscopic Gastritis. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2018. [DOI: 10.22207/jpam.12.3.47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
15
|
Influence of aging in the modulation of epigenetic biomarkers of carcinogenesis after exposure to air pollution. Exp Gerontol 2018; 110:125-132. [DOI: 10.1016/j.exger.2018.05.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 05/11/2018] [Accepted: 05/25/2018] [Indexed: 11/19/2022]
|
16
|
Role of CTCF in DNA damage response. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2018; 780:61-68. [PMID: 31395350 DOI: 10.1016/j.mrrev.2018.02.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 02/20/2018] [Indexed: 12/13/2022]
Abstract
CCCTC-binding factor (CTCF) is a highly conserved, ubiquitously expressed zinc finger protein. CTCF is a multifunctional protein, associated with a number of vital cellular processes such as transcriptional activation, repression, insulation, imprinting and genome organization. Emerging evidence indicates that CTCF is also involved in DNA damage response. In this review, we focus on the newly identified role of CTCF in facilitating DNA double-strand break repair. Due to the large number of cellular processes in which CTCF is involved, factors that functionally affect CTCF could have serious implications on genomic stability. It is becoming increasingly clear that exposure to environmental toxicants could have adverse effects on CTCF functions. Here we discuss the various ways that environmental toxicants could impact CTCF functions and the potential consequences on DNA damage response.
Collapse
|
17
|
Laidlaw KME, Berhan S, Liu S, Silvestri G, Holyoake TL, Frank DA, Aggarwal B, Bonner MY, Perrotti D, Jørgensen HG, Arbiser JL. Cooperation of imipramine blue and tyrosine kinase blockade demonstrates activity against chronic myeloid leukemia. Oncotarget 2018; 7:51651-51664. [PMID: 27438151 PMCID: PMC5239504 DOI: 10.18632/oncotarget.10541] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/30/2016] [Indexed: 01/23/2023] Open
Abstract
The use of tyrosine kinase inhibitors (TKI), including nilotinib, has revolutionized the treatment of chronic myeloid leukemia (CML). However current unmet clinical needs include combating activation of additional survival signaling pathways in persistent leukemia stem cells after long-term TKI therapy. A ubiquitous signaling alteration in cancer, including CML, is activation of reactive oxygen species (ROS) signaling, which may potentiate stem cell activity and mediate resistance to both conventional chemotherapy and targeted inhibitors. We have developed a novel nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitor, imipramine blue (IB) that targets ROS generation. ROS levels are known to be elevated in CML with respect to normal hematopoietic stem/progenitor cells and not corrected by TKI. We demonstrate that IB has additive benefit with nilotinib in inhibiting proliferation, viability, and clonogenic function of TKI-insensitive quiescent CD34+ CML chronic phase (CP) cells while normal CD34+ cells retained their clonogenic capacity in response to this combination therapy in vitro. Mechanistically, the pro-apoptotic activity of IB likely resides in part through its dual ability to block NF-κB and re-activate the tumor suppressor protein phosphatase 2A (PP2A). Combining BCR-ABL1 kinase inhibition with NADPH oxidase blockade may be beneficial in eradication of CML and worthy of further investigation.
Collapse
Affiliation(s)
- Kamilla M E Laidlaw
- Paul O'Gorman Leukemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Gartnavel General Hospital, Glasgow, G12 0ZD, United Kingdom
| | - Samuel Berhan
- Paul O'Gorman Leukemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Gartnavel General Hospital, Glasgow, G12 0ZD, United Kingdom
| | - Suhu Liu
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Giovannino Silvestri
- Department of Medicine, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Tessa L Holyoake
- Paul O'Gorman Leukemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Gartnavel General Hospital, Glasgow, G12 0ZD, United Kingdom
| | - David A Frank
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Bharat Aggarwal
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael Y Bonner
- Department of Dermatology, Emory University School of Medicine, Atlanta, GA 30322, USA.,Atlanta Veterans Administration Hospital, Atlanta, GA 30322, USA
| | - Danilo Perrotti
- Department of Medicine, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Heather G Jørgensen
- Paul O'Gorman Leukemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Gartnavel General Hospital, Glasgow, G12 0ZD, United Kingdom
| | - Jack L Arbiser
- Department of Dermatology, Emory University School of Medicine, Atlanta, GA 30322, USA.,Atlanta Veterans Administration Hospital, Atlanta, GA 30322, USA
| |
Collapse
|
18
|
Canaz E, Kilinc M, Sayar H, Kiran G, Ozyurek E. Lead, selenium and nickel concentrations in epithelial ovarian cancer, borderline ovarian tumor and healthy ovarian tissues. J Trace Elem Med Biol 2017; 43:217-223. [PMID: 28551014 DOI: 10.1016/j.jtemb.2017.05.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 04/13/2017] [Accepted: 05/03/2017] [Indexed: 12/01/2022]
Abstract
OBJECTIVE Wide variation exists in ovarian cancer incidence rates suggesting the importance of environmental factors. Due to increasing environmental pollution, trace elements and heavy metals have drawn attention in studies defining the etiology of cancer, but scant data is available for ovarian cancer. Our aim was to compare the tissue concentrations of lead, selenium and nickel in epithelial ovarian cancer, borderline tumor and healthy ovarian tissues. METHODS The levels of lead, selenium and nickel were estimated using atomic absorption spectrophotometry in formalin-fixed paraffin-embedded tissue samples. Tests were carried out in 20 malignant epithelial ovarian cancer, 15 epithelial borderline tumor and 20 non-neoplastic healthy ovaries. Two samples were collected for borderline tumors, one from papillary projection and one from the smooth surface of cyst wall. RESULTS Pb and Ni concentrations were found to be higher both in malignant and borderline tissues than those in healthy ovaries. Concentrations of Pb and Ni in malignant tissues, borderline papillary projections and capsular tissue samples were not different. Comparison of Se concentrations of malignant, borderline and healthy ovarian tissues did not reveal statistical difference. Studied metal levels were not found to be different in either papillary projection or in cyst wall of the borderline tumors. CONCLUSIONS This study revealed the accumulation of lead and nickel in ovarian tissue is associated with borderline and malignant proliferation of the surface epithelium. Accumulation of these metals in epithelial ovarian cancer and borderline ovarian tumor has not been demonstrated before.
Collapse
Affiliation(s)
- Emel Canaz
- Gynecologic Oncology, Department of Obstetrics and Gynecology, Sisli Hamidiye Etfal Training and Research Hospital, Halaskargazi Cd. Etfal, Sisli, Sk. 34371 Turkey.
| | - Metin Kilinc
- Department of Biochemistry, Kahramanmaras Sutcu Imam University School of Medicine, Kahramanmaras, Turkey
| | - Hamide Sayar
- Department of Pathology, Kahramanmaras Sutcu Imam University School of Medicine, Kahramanmaras, Turkey
| | - Gurkan Kiran
- Department of Obstetrics and Gynecology, Umraniye Training and Research Hospital, Istanbul, Turkey
| | - Eser Ozyurek
- Department of Obstetrics and Gynecology, Bagcilar Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
19
|
Prasad R, Singh T, Katiyar SK. Honokiol inhibits ultraviolet radiation-induced immunosuppression through inhibition of ultraviolet-induced inflammation and DNA hypermethylation in mouse skin. Sci Rep 2017; 7:1657. [PMID: 28490739 PMCID: PMC5431968 DOI: 10.1038/s41598-017-01774-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 03/30/2017] [Indexed: 01/10/2023] Open
Abstract
Ultraviolet (UV) radiation exposure induces immunosuppression, which contributes to the development of cutaneous malignancies. We investigated the effects of honokiol, a phytochemical found in plants of the genus Magnolia, on UVB-induced immunosuppression using contact hypersensitivity (CHS) as a model in C3H/HeN mice. Topical application of honokiol (0.5 and 1.0 mg/cm2 skin area) had a significant preventive effect on UVB-induced suppression of the CHS response. The inflammatory mediators, COX-2 and PGE2, played a key role in this effect, as indicated by honokiol inhibition of cyclooxygenase-2 (COX-2) expression and PGE2 production in the UVB-exposed skin. Honokiol application also inhibited UVB-induced DNA hypermethylation and its elevation of the levels of TET enzyme, which is responsible for DNA demethylation in UVB-exposed skin. This was consistent with the restoration of the CHS response in mice treated with the DNA demethylating agent, 5-aza-2'-deoxycytidine, after UVB exposure. There was no significant difference in the levels of inhibition of UVB-induced immunosuppression amongst mice that were treated topically with available anti-cancer drugs (imiquimod and 5-fluorouracil). This study is the first to show that honokiol has the ability to inhibit UVB-induced immunosuppression in preclinical model and, thus, has potential for use as a chemopreventive strategy for UVB radiation-induced malignancies.
Collapse
Affiliation(s)
- Ram Prasad
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA
- Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA
| | - Tripti Singh
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Santosh K Katiyar
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA.
- Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA.
- Environmental Health Sciences, University of Alabama at Birmingham, Birmingham, AL, USA.
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
20
|
Salemi R, Marconi A, Di Salvatore V, Franco S, Rapisarda V, Libra M. Epigenetic alterations and occupational exposure to benzene, fibers, and heavy metals associated with tumor development. Mol Med Rep 2017; 15:3366-3371. [DOI: 10.3892/mmr.2017.6383] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 02/16/2017] [Indexed: 11/05/2022] Open
|
21
|
Franken C, Koppen G, Lambrechts N, Govarts E, Bruckers L, Den Hond E, Loots I, Nelen V, Sioen I, Nawrot TS, Baeyens W, Van Larebeke N, Boonen F, Ooms D, Wevers M, Jacobs G, Covaci A, Schettgen T, Schoeters G. Environmental exposure to human carcinogens in teenagers and the association with DNA damage. ENVIRONMENTAL RESEARCH 2017; 152:165-174. [PMID: 27771571 DOI: 10.1016/j.envres.2016.10.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 10/10/2016] [Accepted: 10/13/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND We investigated whether human environmental exposure to chemicals that are labeled as (potential) carcinogens leads to increased (oxidative) damage to DNA in adolescents. MATERIAL AND METHODS Six hundred 14-15-year-old youngsters were recruited all over Flanders (Belgium) and in two areas with important industrial activities. DNA damage was assessed by alkaline and formamidopyrimidine DNA glycosylase (Fpg) modified comet assays in peripheral blood cells and analysis of urinary 8-hydroxydeoxyguanosine (8-OHdG) levels. Personal exposure to potentially carcinogenic compounds was measured in urine, namely: chromium, cadmium, nickel, 1-hydroxypyrene as a proxy for exposure to other carcinogenic polycyclic aromatic hydrocarbons (PAHs), t,t-muconic acid as a metabolite of benzene, 2,5-dichlorophenol (2,5-DCP), organophosphate pesticide metabolites, and di(2-ethylhexyl) phthalate (DEHP) metabolites. In blood, arsenic, polychlorinated biphenyl (PCB) congeners 118 and 156, hexachlorobenzene (HCB), dichlorodiphenyltrichloroethane (DDT) and perfluorooctanoic acid (PFOA) were analyzed. Levels of methylmercury (MeHg) were measured in hair. Multiple linear regression models were used to establish exposure-response relationships. RESULTS Biomarkers of exposure to PAHs and urinary chromium were associated with higher levels of both 8-OHdG in urine and DNA damage detected by the alkaline comet assay. Concentrations of 8-OHdG in urine increased in relation with increasing concentrations of urinary t,t-muconic acid, cadmium, nickel, 2,5-DCP, and DEHP metabolites. Increased concentrations of PFOA in blood were associated with higher levels of DNA damage measured by the alkaline comet assay, whereas DDT was associated in the same direction with the Fpg-modified comet assay. Inverse associations were observed between blood arsenic, hair MeHg, PCB 156 and HCB, and urinary 8-OHdG. The latter exposure biomarkers were also associated with higher fish intake. Urinary nickel and t,t-muconic acid were inversely associated with the alkaline comet assay. CONCLUSION This cross-sectional study found associations between current environmental exposure to (potential) human carcinogens in 14-15-year-old Flemish adolescents and short-term (oxidative) damage to DNA. Prospective follow-up will be required to investigate whether long-term effects may occur due to complex environmental exposures.
Collapse
Affiliation(s)
- Carmen Franken
- Flemish Institute for Technological Research (VITO), Mol, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| | - Gudrun Koppen
- Flemish Institute for Technological Research (VITO), Mol, Belgium
| | | | - Eva Govarts
- Flemish Institute for Technological Research (VITO), Mol, Belgium
| | - Liesbeth Bruckers
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Hasselt University, Hasselt, Belgium
| | - Elly Den Hond
- Flemish Institute for Technological Research (VITO), Mol, Belgium
| | - Ilse Loots
- Political and Social Sciences, University of Antwerp, Antwerp, Belgium
| | - Vera Nelen
- Provincial Institute for Hygiene, Antwerp, Belgium
| | - Isabelle Sioen
- Department of Public Health, Ghent University, Ghent, Belgium; Department of Food Safety and Food Quality, Ghent University, Ghent, Belgium
| | - Tim S Nawrot
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium; Department of Public Health & Primary Care, Leuven University, Leuven, Belgium
| | - Willy Baeyens
- Department of Analytical and Environmental Chemistry, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nicolas Van Larebeke
- Department of Analytical and Environmental Chemistry, Vrije Universiteit Brussel, Brussels, Belgium; Department of Radiotherapy and Experimental Cancerology, Ghent University, Ghent, Belgium
| | - Francis Boonen
- Flemish Institute for Technological Research (VITO), Mol, Belgium
| | - Daniëlla Ooms
- Flemish Institute for Technological Research (VITO), Mol, Belgium
| | - Mai Wevers
- Flemish Institute for Technological Research (VITO), Mol, Belgium
| | - Griet Jacobs
- Flemish Institute for Technological Research (VITO), Mol, Belgium
| | - Adrian Covaci
- Toxicological Centre, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Thomas Schettgen
- Department of Occupational and Social Medicine, RWTH Aachen University, Aachen, Germany
| | - Greet Schoeters
- Flemish Institute for Technological Research (VITO), Mol, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; University of Southern Denmark, Institute of Public Health, Department of Environmental Medicine, Odense, Denmark
| |
Collapse
|
22
|
Zeng L, Ma H, Pan S, You J, Zhang G, Yu Z, Sheng G, Fu J. LINE-1 gene hypomethylation and p16 gene hypermethylation in HepG2 cells induced by low-dose and long-term triclosan exposure: The role of hydroxyl group. Toxicol In Vitro 2016; 34:35-44. [DOI: 10.1016/j.tiv.2016.03.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 02/20/2016] [Accepted: 03/06/2016] [Indexed: 12/18/2022]
|
23
|
Jha HC, Banerjee S, Robertson ES. The Role of Gammaherpesviruses in Cancer Pathogenesis. Pathogens 2016; 5:pathogens5010018. [PMID: 26861404 PMCID: PMC4810139 DOI: 10.3390/pathogens5010018] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 01/27/2016] [Indexed: 12/15/2022] Open
Abstract
Worldwide, one fifth of cancers in the population are associated with viral infections. Among them, gammaherpesvirus, specifically HHV4 (EBV) and HHV8 (KSHV), are two oncogenic viral agents associated with a large number of human malignancies. In this review, we summarize the current understanding of the molecular mechanisms related to EBV and KSHV infection and their ability to induce cellular transformation. We describe their strategies for manipulating major cellular systems through the utilization of cell cycle, apoptosis, immune modulation, epigenetic modification, and altered signal transduction pathways, including NF-kB, Notch, Wnt, MAPK, TLR, etc. We also discuss the important EBV latent antigens, namely EBNA1, EBNA2, EBNA3’s and LMP’s, which are important for targeting these major cellular pathways. KSHV infection progresses through the engagement of the activities of the major latent proteins LANA, v-FLIP and v-Cyclin, and the lytic replication and transcription activator (RTA). This review is a current, comprehensive approach that describes an in-depth understanding of gammaherpes viral encoded gene manipulation of the host system through targeting important biological processes in viral-associated cancers.
Collapse
Affiliation(s)
- Hem Chandra Jha
- Department of Microbiology and Tumor Virology Program, Abramson Comprehensive Cancer Center, Perelman School of Medicine at the University of Pennsylvania, 201E Johnson Pavilion, 3610, Hamilton Walk, Philadelphia, PA 19104, USA.
| | - Shuvomoy Banerjee
- Department of Microbiology and Tumor Virology Program, Abramson Comprehensive Cancer Center, Perelman School of Medicine at the University of Pennsylvania, 201E Johnson Pavilion, 3610, Hamilton Walk, Philadelphia, PA 19104, USA.
| | - Erle S Robertson
- Department of Microbiology and Tumor Virology Program, Abramson Comprehensive Cancer Center, Perelman School of Medicine at the University of Pennsylvania, 201E Johnson Pavilion, 3610, Hamilton Walk, Philadelphia, PA 19104, USA.
| |
Collapse
|
24
|
Histone Methylation in Nickel-Smelting Industrial Workers. PLoS One 2015; 10:e0140339. [PMID: 26474320 PMCID: PMC4608576 DOI: 10.1371/journal.pone.0140339] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 09/24/2015] [Indexed: 11/23/2022] Open
Abstract
Background Nickel is an essential trace metal naturally found in the environment. It is also common in occupational settings, where it associates with various levels of both occupational and nonoccupational exposure In vitro studies have shown that nickel exposure can lead to intracellular accumulation of Ni2+, which has been associated with global decreases in DNA methylation, increases in chromatin condensation, reductions in H3K9me2, and elevated levels of H3K4me3. Histone modifications play an important role in modulating chromatin structure and gene expression. For example, tri-methylation of histone H3k4 has been found to be associated with transcriptional activation, and tri-methylation of H3k27 has been found to be associated with transcriptional repression. Aberrant histone modifications have been found to be associated with various human diseases, including cancer. The purpose of this work was to identify biomarkers for populations with occupational nickel exposure and to examine the relationship between histone methylation and nickel exposure. This may provide a scientific indicator of early health impairment and facilitate exploration of the molecular mechanism underlying cancer pathogenesis. Methods One hundred and forty subjects with occupational exposure to Ni and 140 referents were recruited. H3K4 and H3K27 trimethylation levels were measured in subjects’ blood cells. Results H3K4me3 levels were found to be higher in nickel smelting workers (47.24±20.85) than in office workers (22.65±8.81; P = 0.000), while the opposite was found for levels of H3K27me3(nickel smelting workers, 13.88± 4.23; office workers, 20.67± 5.96; P = 0.000). H3K4me3 was positively (r = 0.267, P = 0.001) and H3K27 was negatively (r = -0.684, P = 0.000) associated with age and length of service in smelting workers. Conclusion This study indicated that occupational exposure to Ni is associated with alterations in levels of histone modification.
Collapse
|
25
|
Scientific Opinion on the risks to public health related to the presence of nickel in food and drinking water. EFSA J 2015. [DOI: 10.2903/j.efsa.2015.4002] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
26
|
Crujeiras AB, Casanueva FF. Obesity and the reproductive system disorders: epigenetics as a potential bridge. Hum Reprod Update 2014; 21:249-61. [DOI: 10.1093/humupd/dmu060] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
27
|
Brocato J, Costa M. 10th NTES Conference: Nickel and Arsenic Compounds Alter the Epigenome of Peripheral Blood Mononuclear Cells. J Trace Elem Med Biol 2014; 31:209-13. [PMID: 24837610 PMCID: PMC4201979 DOI: 10.1016/j.jtemb.2014.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 03/31/2014] [Accepted: 04/07/2014] [Indexed: 11/18/2022]
Abstract
The mechanisms that underlie metal carcinogenesis are the subject of intense investigation; however, data from in vitro and in vivo studies are starting to piece together a story that implicates epigenetics as a key player. Data from our lab has shown that nickel compounds inhibit dioxygenase enzymes by displacing iron in the active site. Arsenic is hypothesized to inhibit these enzymes by diminishing ascorbate levels--an important co-factor for dioxygenases. Inhibition of histone demethylase dioxygenases can increase histone methylation levels, which also may affect gene expression. Recently, our lab conducted a series of investigations in human subjects exposed to high levels of nickel or arsenic compounds. Global levels of histone modifications in peripheral blood mononuclear cells (PBMCs) from exposed subjects were compared to low environmentally exposed controls. Results showed that nickel increased H3K4me3 and decreased H3K9me2 globally. Arsenic increased H3K9me2 and decreased H3K9ac globally. Other histone modifications affected by arsenic were sex-dependent. Nickel affected the expression of 2756 genes in human PBMCs and many of the genes were involved in immune and carcinogenic pathways. This review will describe data from our lab that demonstrates for the first time that nickel and arsenic compounds affect global levels of histone modifications and gene expression in exposed human populations.
Collapse
Affiliation(s)
- Jason Brocato
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 57 Old Forge Road, NY 10987, USA
| | - Max Costa
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 57 Old Forge Road, NY 10987, USA.
| |
Collapse
|
28
|
Denat L, Kadekaro AL, Marrot L, Leachman SA, Abdel-Malek ZA. Melanocytes as instigators and victims of oxidative stress. J Invest Dermatol 2014; 134:1512-1518. [PMID: 24573173 PMCID: PMC4418514 DOI: 10.1038/jid.2014.65] [Citation(s) in RCA: 261] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 01/08/2014] [Accepted: 01/15/2014] [Indexed: 12/30/2022]
Abstract
Epidermal melanocytes are particularly vulnerable to oxidative stress owing to the pro-oxidant state generated during melanin synthesis, and to the intrinsic antioxidant defenses that are compromised in pathologic conditions. Melanoma is thought to be oxidative stress driven, and melanocyte death in vitiligo is thought to be instigated by a highly pro-oxidant state in the epidermis. We review the current knowledge about melanin and the redox state of melanocytes, how paracrine factors help counteract oxidative stress, the role of oxidative stress in melanoma initiation and progression and in melanocyte death in vitiligo, and how this knowledge can be harnessed for melanoma and vitiligo treatment.
Collapse
Affiliation(s)
- Laurence Denat
- L'OREAL Research and Innovation, Aulnay-sous-Bois, France
| | - Ana L Kadekaro
- Department of Dermatology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Laurent Marrot
- L'OREAL Research and Innovation, Aulnay-sous-Bois, France
| | - Sancy A Leachman
- Department of Dermatology, Oregon Health Sciences University, Portland, Oregon, USA
| | | |
Collapse
|
29
|
Zhang J, Zhou Y, Wu YJ, Li MJ, Wang RJ, Huang SQ, Gao RR, Ma L, Shi HJ, Zhang J. Hyper-methylated miR-203 dysregulates ABL1 and contributes to the nickel-induced tumorigenesis. Toxicol Lett 2013; 223:42-51. [PMID: 23968727 DOI: 10.1016/j.toxlet.2013.08.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 08/07/2013] [Accepted: 08/09/2013] [Indexed: 10/26/2022]
Abstract
Nickel compounds have been found to be carcinogenic based upon epidemiological, animal and cell culture studies. Previous studies suggest that epigenetic mechanisms play a role in Nickel-induced carcinogenesis such as DNA methylation and histone modification. In this study, we investigated the role of microRNAs (miRNAs) in nickel-induced carcinogenesis. The expression of several miRNAs which may function as tumor suppressor genes revealed a strong downregulation of miR-203 in Ni3S2-transformed 16HBE cells (NSTCs). Meanwhile, we observed hypermethylation of CpGs in miR-203 promoter and first exon area, and proved that the hyper-methylated miR-203 was involved in the Nickel-induced tumorigenesis. Moreover, we identified that miR-203 may suppress the tumorigenesis at least in part through negatively regulating its target gene ABL1. Our findings indicate that DNA methylation-associated silencing of tumor suppressor miRNAs contributes to the development of Nickel-induced cancer.
Collapse
Affiliation(s)
- Jing Zhang
- Translational Center for Stem Cell Research, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; School of Life Science and Technology, Tongji University, Shanghai 200092, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Toxicogenomic approaches for understanding molecular mechanisms of heavy metal mutagenicity and carcinogenicity. Int J Hyg Environ Health 2013; 216:587-98. [PMID: 23540489 DOI: 10.1016/j.ijheh.2013.02.010] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 02/26/2013] [Accepted: 02/26/2013] [Indexed: 12/24/2022]
Abstract
Heavy metals that are harmful to humans include arsenic, cadmium, chromium, lead, mercury, and nickel. Some metals or their related compounds may even cause cancer. However, the mechanism underlying heavy metal-induced cancer remains unclear. Increasing data show a link between heavy metal exposure and aberrant changes in both genetic and epigenetic factors via non-targeted multiple toxicogenomic technologies of the transcriptome, proteome, metabolome, and epigenome. These modifications due to heavy metal exposure might provide a better understanding of environmental disorders. Such informative changes following heavy metal exposure might also be useful for screening of biomarker-monitored exposure to environmental pollutants and/or predicting the risk of disease. We summarize advances in high-throughput toxicogenomic-based technologies and studies related to exposure to individual heavy metal and/or mixtures and propose the underlying mechanism of action and toxicant signatures. Integrative multi-level expression analysis of the toxicity of heavy metals via system toxicology-based methodologies combined with statistical and computational tools might clarify the biological pathways involved in carcinogenic processes. Although standard in vitro and in vivo endpoint testing of mutagenicity and carcinogenicity are considered a complementary approach linked to disease, we also suggest that further evaluation of prominent biomarkers reflecting effects, responses, and disease susceptibility might be diagnostic. Furthermore, we discuss challenges in toxicogenomic applications for toxicological studies of metal mixtures and epidemiological research. Taken together, this review presents toxicogenomic data that will be useful for improvement of the knowledge of carcinogenesis and the development of better strategies for health risk assessment.
Collapse
|
31
|
Abstract
This review focuses on the impact of nickel on human health. In particular, the dual nature of nickel as an essential as well as toxic element in nature is described, and the main forms of nickel that can come in contact with living systems from natural sources and anthropogenic activities are discussed. Concomitantly, the main routes of nickel uptake and transport in humans are covered, and the potential dangers that nickel exposure can represent for health are described. In particular, the insurgence of nickel-derived allergies, nickel-induced carcinogenesis as well as infectious diseases caused by human pathogens that rely on nickel-based enzymes to colonize the host are reviewed at different levels, from their macroscopic aspects on human health to the molecular mechanisms underlying these points. Finally, the importance of nickel as a beneficial element for human health, especially being essential for microorganisms that colonize the human guts, is examined.
Collapse
Affiliation(s)
- Barbara Zambelli
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy,
| | | |
Collapse
|
32
|
Muñoz A, Costa M. Elucidating the mechanisms of nickel compound uptake: a review of particulate and nano-nickel endocytosis and toxicity. Toxicol Appl Pharmacol 2012; 260:1-16. [PMID: 22206756 PMCID: PMC3306469 DOI: 10.1016/j.taap.2011.12.014] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 12/08/2011] [Accepted: 12/12/2011] [Indexed: 10/14/2022]
Abstract
Nickel (Ni) is a worldwide pollutant and contaminant that humans are exposed to through various avenues resulting in multiple toxic responses - most alarming is its clear carcinogenic nature. A variety of particulate Ni compounds persist in the environment and can be distinguished by characteristics such as solubility, structure, and surface charge. These characteristics influence cellular uptake and toxicity. Some particulate forms of Ni are carcinogenic and are directly and rapidly endocytized by cells. A series of studies conducted in the 1980s observed this process, and we have reanalyzed the results of these studies to help elucidate the molecular mechanism of particulate Ni uptake. Originally the process of uptake observed was described as phagocytosis, however in the context of recent research we hypothesize that the process is macropinocytosis and/or clathrin mediated endocytosis. Primary considerations in determining the route of uptake here include calcium dependence, particle size, and inhibition through temperature and pharmacological approaches. Particle characteristics that influenced uptake include size, charge, surface characteristics, and structure. This discussion is relevant in the context of nanoparticle studies and the emerging interest in nano-nickel (nano-Ni), where toxicity assessments require a clear understanding of the parameters of particulate uptake and where establishment of such parameters is often obscured through inconsistencies across experimental systems. In this regard, this review aims to carefully document one system (particulate nickel compound uptake) and characterize its properties.
Collapse
Affiliation(s)
- Alexandra Muñoz
- New York University School of Medicine, Nelson Institute of Environmental Medicine, 57 Old Forge Road, NY 10987
| | - Max Costa
- New York University School of Medicine, Nelson Institute of Environmental Medicine, 57 Old Forge Road, NY 10987
| |
Collapse
|
33
|
Cheng TF, Choudhuri S, Muldoon-Jacobs K. Epigenetic targets of some toxicologically relevant metals: a review of the literature. J Appl Toxicol 2012; 32:643-53. [DOI: 10.1002/jat.2717] [Citation(s) in RCA: 136] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 12/13/2011] [Accepted: 12/13/2011] [Indexed: 12/13/2022]
Affiliation(s)
- Tsu-Fan Cheng
- US Food and Drug Administration; Center for Food Safety and Applied Nutrition, Office of Food Additive Safety, Division of Food Contact Notification; College Park; MD; USA
| | - Supratim Choudhuri
- US Food and Drug Administration; Center for Food Safety and Applied Nutrition, Office of Food Additive Safety, Division of Biotechnology and GRAS Notice Review; College Park; MD; USA
| | - Kristi Muldoon-Jacobs
- US Food and Drug Administration; Center for Food Safety and Applied Nutrition, Office of Food Additive Safety, Division of Food Contact Notification; College Park; MD; USA
| |
Collapse
|
34
|
Yang P, Ma J, Zhang B, Duan H, He Z, Zeng J, Zeng X, Li D, Wang Q, Xiao Y, Liu C, Xiao Q, Chen L, Zhu X, Xing X, Li Z, Zhang S, Zhang Z, Ma L, Wang E, Zhuang Z, Zheng Y, Chen W. CpG site-specific hypermethylation of p16INK4α in peripheral blood lymphocytes of PAH-exposed workers. Cancer Epidemiol Biomarkers Prev 2011; 21:182-90. [PMID: 22028397 DOI: 10.1158/1055-9965.epi-11-0784] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Sufficient epidemiologic evidence shows an etiologic link between polycyclic aromatic hydrocarbons (PAH) exposure and lung cancer risk. While the genetic modifications have been found in PAH-exposed population, it is unclear whether gene-specific methylation involves in the process of PAH-associated biologic consequence. METHODS Sixty-nine PAH-exposed workers and 59 control subjects were recruited. Using bisulfite sequencing, we examined the methylation status of p16(INK4α) promoter in peripheral blood lymphocytes (PBL) from PAH-exposed workers and in benzo(a)pyrene (BaP)-transformed human bronchial epithelial (HBE) cells. The relationships between p16(INK4α) methylation and the level of urinary 1-hydroxypyrene (1-OHP) or the frequency of cytokinesis block micronucleus (CBMN) were analyzed. RESULTS Compared with the control group, PAH-exposed workers exhibited higher levels of urinary 1-OHP (10.62 vs. 2.52 μg/L), p16(INK4α) methylation (7.95% vs. 1.14% for 22 "hot" CpG sites), and CBMN (7.28% vs. 2.92%) in PBLs. p16(INK4α) hypermethylation in PAH-exposed workers exhibited CpG site specificity. Among the 35 CpG sites we analyzed, 22 were significantly hypermethylated. These 22 hypermethylated CpG sites were positively correlated to levels of urinary 1-OHP and CBMN in PBLs. Moreover, the hypermethylation and suppression of p16 expression was also found in BaP-transformed HBER cells. CONCLUSION PAH exposure induced CpG site-specific hypermethylation of p16(INK4α) gene. The degree of p16(INK4α) methylation was associated with the levels of DNA damage and internal exposure. IMPACT p16(INK4α) hypermethylation might be an essential biomarker for the exposure to PAHs and for early diagnosis of cancer.
Collapse
Affiliation(s)
- Ping Yang
- Department of Toxicology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Okazaki R, Ootsuyama A, Kakihara H, Mabuchi Y, Matsuzaki Y, Michikawa Y, Imai T, Norimura T. Dynamics of Delayed p53 Mutations in Mice Given Whole-Body Irradiation at 8 Weeks. Int J Radiat Oncol Biol Phys 2011; 79:247-54. [DOI: 10.1016/j.ijrobp.2010.07.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 07/05/2010] [Accepted: 07/07/2010] [Indexed: 01/11/2023]
|
36
|
Chen H, Kluz T, Zhang R, Costa M. Hypoxia and nickel inhibit histone demethylase JMJD1A and repress Spry2 expression in human bronchial epithelial BEAS-2B cells. Carcinogenesis 2010; 31:2136-44. [PMID: 20881000 DOI: 10.1093/carcin/bgq197] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Epigenetic silencing of tumor suppressor genes commonly occurs in human cancers via increasing DNA methylation and repressive histone modifications at gene promoters. However, little is known about how pathogenic environmental factors contribute to cancer development by affecting epigenetic regulatory mechanisms. Previously, we reported that both hypoxia and nickel (an environmental carcinogen) increased global histone H3 lysine 9 methylation in cells through inhibiting a novel class of iron- and α-ketoglutarate-dependent histone demethylases. Here, we investigated whether inhibition of histone demethylase JMJD1A by hypoxia and nickel could lead to repression/silencing of JMJD1A-targeted gene(s). By using Affymetrix GeneChip and ChIP-on-chip technologies, we identified Spry2 gene, a key regulator of receptor tyrosine kinase/extracellular signal-regulated kinase (ERK) signaling, as one of the JMJD1A-targeted genes in human bronchial epithelial BEAS-2B cells. Both hypoxia and nickel exposure increased the level of H3K9me2 at the Spry2 promoter by inhibiting JMJD1A, which probably led to a decreased expression of Spry2 in BEAS-2B cells. Repression of Spry2 potentiated the nickel-induced ERK phosphorylation, and forced expression of Spry2 in BEAS-2B cells decreased the nickel-induced ERK phosphorylation and significantly suppressed nickel-induced anchorage-independent growth. Taken together, our results suggest that histone demethylases could be targets of environmental carcinogens and their inhibition may lead to altered gene expression and eventually carcinogenesis.
Collapse
Affiliation(s)
- Haobin Chen
- Department of Environmental Medicine, New York University of School of Medicine, 550 First Avenue, New York, NY 10016, USA.
| | | | | | | |
Collapse
|
37
|
Wettergren Y, Odin E, Carlsson G, Gustavsson B. MTHFR, MTR, and MTRR polymorphisms in relation to p16INK4A hypermethylation in mucosa of patients with colorectal cancer. Mol Med 2010; 16:425-32. [PMID: 20549016 DOI: 10.2119/molmed.2009.00156] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2009] [Accepted: 06/10/2010] [Indexed: 12/17/2022] Open
Abstract
We recently analyzed the hypermethylation status of the p16INK4a (p16) gene promoter in normal-appearing mucosa obtained from patients with colorectal cancer. Hypermethylation of p16 was associated with reduced survival of these patients. In the present study, germ line polymorphisms in the folate- and methyl-associated genes, methylenetetrahydrofolate reductase (MTHFR), methionine synthase (MTR) and methionine synthase reductase (MTRR), were analyzed in the same patient cohort to find a possible link between these genetic variants and p16 hypermethylation. Genomic DNA was extracted from blood of patients (n = 181) and controls (n = 300). Genotype analyses were run on an ABI PRISM(®) 7900HT sequence-detection system (Applied Biosystems), using real-time polymerase chain reaction and TaqMan chemistry. The results showed that the genotype distributions of the patient and control groups were similar. No significant differences in cancer-specific or disease-free survival of stage I-III patients according to polymorphic variants were detected, nor were any differences in cancer-specific or disease-free survival detected when patients were subgrouped according to the MTHFR or MTR genotype groups and dichotomized by p16 hypermethylation status in mucosa. However, patients with the MTRR 66 AA/AG genotypes were found to have a significantly worse cancer-specific survival when the mucosa were positive, compared with negative, for p16 hypermethylation (hazard ratio 2.7; 95% confidence interval 1.2-6.4; P = 0.023). In contrast, there was no difference in survival among patients with the MTRR 66 GG genotype stratified by p16 hypermethylation status. These results indicate a relationship between genetic germ-line variants of the MTRR gene and p16 hypermethylation in mucosa, which may affect the clinical outcome of patients with colorectal cancer.
Collapse
Affiliation(s)
- Yvonne Wettergren
- Department of General Surgery, University of Gothenburg, Sahlgrenska University Hospital/Ostra, Gothenburg, Sweden.
| | | | | | | |
Collapse
|
38
|
Nunes AM, Zavitsanos K, Del Conte R, Malandrinos G, Hadjiliadis N. The Possible Role of 94−125 Peptide Fragment of Histone H2B in Nickel-Induced Carcinogenesis. Inorg Chem 2010; 49:5658-68. [DOI: 10.1021/ic1005665] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Ana Mónica Nunes
- Department of Chemistry, University of Ioannina, 45110 Ioannina, Greece
| | - Kimon Zavitsanos
- Department of Chemistry, University of Ioannina, 45110 Ioannina, Greece
| | - Rebecca Del Conte
- Magnetic Resonance Center, University of Florence, Via L. Sacconi 6, 0019, Sesto Fiorentino, Italy
| | | | - Nick Hadjiliadis
- Department of Chemistry, University of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
39
|
Chen H, Giri NC, Zhang R, Yamane K, Zhang Y, Maroney M, Costa M. Nickel ions inhibit histone demethylase JMJD1A and DNA repair enzyme ABH2 by replacing the ferrous iron in the catalytic centers. J Biol Chem 2010; 285:7374-83. [PMID: 20042601 PMCID: PMC2844186 DOI: 10.1074/jbc.m109.058503] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2009] [Revised: 12/23/2009] [Indexed: 01/20/2023] Open
Abstract
Iron- and 2-oxoglutarate-dependent dioxygenases are a diverse family of non-heme iron enzymes that catalyze various important oxidations in cells. A key structural motif of these dioxygenases is a facial triad of 2-histidines-1-carboxylate that coordinates the Fe(II) at the catalytic site. Using histone demethylase JMJD1A and DNA repair enzyme ABH2 as examples, we show that this family of dioxygenases is highly sensitive to inhibition by carcinogenic nickel ions. We find that, with iron, the 50% inhibitory concentrations of nickel (IC(50) [Ni(II)]) are 25 microm for JMJD1A and 7.5 microm for ABH2. Without iron, JMJD1A is 10 times more sensitive to nickel inhibition with an IC(50) [Ni(II)] of 2.5 microm, and approximately one molecule of Ni(II) inhibits one molecule of JMJD1A, suggesting that nickel causes inhibition by replacing the iron. Furthermore, nickel-bound JMJD1A is not reactivated by excessive iron even up to a 2 mm concentration. Using x-ray absorption spectroscopy, we demonstrate that nickel binds to the same site in ABH2 as iron, and replacement of the iron by nickel does not prevent the binding of the cofactor 2-oxoglutarate. Finally, we show that nickel ions target and inhibit JMJD1A in intact cells, and disruption of the iron-binding site decreases binding of nickel ions to ABH2 in intact cells. Together, our results reveal that the members of this dioxygenase family are specific targets for nickel ions in cells. Inhibition of these dioxygenases by nickel is likely to have widespread impacts on cells (e.g. impaired epigenetic programs and DNA repair) and may eventually lead to cancer development.
Collapse
Affiliation(s)
- Haobin Chen
- From the Department of Environmental Medicine, New York University of School of Medicine, New York, New York 10016
| | - Nitai Charan Giri
- the Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01002, and
| | - Ronghe Zhang
- From the Department of Environmental Medicine, New York University of School of Medicine, New York, New York 10016
| | - Kenichi Yamane
- the Department of Biochemistry and Biophysics, Howard Hughes Medical Institute, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Yi Zhang
- the Department of Biochemistry and Biophysics, Howard Hughes Medical Institute, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Michael Maroney
- the Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01002, and
| | - Max Costa
- From the Department of Environmental Medicine, New York University of School of Medicine, New York, New York 10016
| |
Collapse
|
40
|
|
41
|
van Kruijsdijk RCM, van der Wall E, Visseren FLJ. Obesity and cancer: the role of dysfunctional adipose tissue. Cancer Epidemiol Biomarkers Prev 2009; 18:2569-78. [PMID: 19755644 DOI: 10.1158/1055-9965.epi-09-0372] [Citation(s) in RCA: 517] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Overweight and obesity are health problems of epidemic proportions, increasing the risk not only of cardiovascular disease and type 2 diabetes mellitus but also of various types of cancer. Obesity is strongly associated with changes in the physiological function of adipose tissue, leading to insulin resistance, chronic inflammation, and altered secretion of adipokines. Several of these factors, such as insulin resistance, increased levels of leptin, plasminogen activator inhibitor-1, and endogenous sex steroids, decreased levels of adiponectin, and chronic inflammation, are involved in carcinogenesis and cancer progression. This article reviews these mechanisms, focusing on adipose tissue dysfunction as a unifying causal factor. Although understanding of the link between obesity and cancer might provide therapeutic targets, preventing overweight and obesity still remains number one priority.
Collapse
Affiliation(s)
- Rob C M van Kruijsdijk
- Department of Vascular Medicine, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands
| | | | | |
Collapse
|
42
|
Sanchez VC, Pietruska JR, Miselis NR, Hurt RH, Kane AB. Biopersistence and potential adverse health impacts of fibrous nanomaterials: what have we learned from asbestos? WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2009; 1:511-29. [PMID: 20049814 PMCID: PMC2864601 DOI: 10.1002/wnan.41] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Human diseases associated with exposure to asbestos fibers include pleural fibrosis and plaques, pulmonary fibrosis (asbestosis), lung cancer, and diffuse malignant mesothelioma. The critical determinants of fiber bioactivity and toxicity include not only fiber dimensions, but also shape, surface reactivity, crystallinity, chemical composition, and presence of transition metals. Depending on their size and dimensions, inhaled fibers can penetrate the respiratory tract to the distal airways and into the alveolar spaces. Fibers can be cleared by several mechanisms, including the mucociliary escalator, engulfment, and removal by macrophages, or through splitting and chemical modification. Biopersistence of long asbestos fibers can lead to inflammation, granuloma formation, fibrosis, and cancer. Exposure to synthetic carbon nanomaterials, including carbon nanofibers and carbon nanotubes (CNTs), is considered a potential health hazard because of their physical similarities with asbestos fibers. Respiratory exposure to CNTs can produce an inflammatory response, diffuse interstitial fibrosis, and formation of fibrotic granulomas similar to that observed in asbestos-exposed animals and humans. Given the known cytotoxic and carcinogenic properties of asbestos fibers, toxicity of fibrous nanomaterials is a topic of intense study. The mechanisms of nanomaterial toxicity remain to be fully elucidated, but recent evidence suggests points of similarity with asbestos fibers, including a role for generation of reactive oxygen species, oxidative stress, and genotoxicity. Considering the rapid increase in production and use of fibrous nanomaterials, it is imperative to gain a thorough understanding of their biologic activity to avoid the human health catastrophe that has resulted from widespread use of asbestos fibers.
Collapse
Affiliation(s)
- Vanesa C. Sanchez
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Jodie R. Pietruska
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Nathan R. Miselis
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Robert H. Hurt
- Division of Engineering, Brown University, Providence, RI, USA
| | - Agnes B. Kane
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| |
Collapse
|
43
|
Reactive oxygen species and mitogen-activated protein kinase activation in the development of Burkitt's lymphoma in Crohn disease. J Clin Gastroenterol 2009; 43:598-9. [PMID: 19034040 DOI: 10.1097/mcg.0b013e3181734a41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
|
44
|
Abstract
PURPOSE OF REVIEW Epigenetics investigates heritable changes in gene expression occurring without changes in DNA sequence. Several epigenetic mechanisms, including DNA methylation, histone modifications, and microRNA expression, can change genome function under exogenous influence. Here, we review current evidence indicating that epigenetic alterations mediate toxicity from environmental chemicals. RECENT FINDINGS In-vitro, animal, and human investigations have identified several classes of environmental chemicals that modify epigenetic marks, including metals (cadmium, arsenic, nickel, chromium, and methylmercury), peroxisome proliferators (trichloroethylene, dichloroacetic acid, and TCA), air pollutants (particulate matter, black carbon, and benzene), and endocrine-disrupting/reproductive toxicants (diethylstilbestrol, bisphenol A, persistent organic pollutants, and dioxin). Most studies conducted so far have been centered on DNA methylation, whereas only a few investigations have studied environmental chemicals in relation to histone modifications and microRNA. SUMMARY For several exposures, it has been proved that chemicals can alter epigenetic marks, and that the same or similar epigenetic alterations can be found in patients with the disease of concern or in diseased tissues. Future prospective investigations are needed to determine whether exposed individuals develop epigenetic alterations over time and, in turn, which such alterations increase the risk of disease. Also, further research is needed to determine whether environmental epigenetic changes are transmitted transgenerationally.
Collapse
Affiliation(s)
- Andrea Baccarelli
- Laboratory of Environmental Epigenetics, Center of Molecular and Genetic Epidemiology, Department of Environmental and Occupational Health, University of Milan, Via San Barnaba 8, Milan, Italy.
| | | |
Collapse
|
45
|
Li Q, Suen TC, Sun H, Arita A, Costa M. Nickel compounds induce apoptosis in human bronchial epithelial Beas-2B cells by activation of c-Myc through ERK pathway. Toxicol Appl Pharmacol 2009; 235:191-8. [PMID: 19135467 PMCID: PMC2657554 DOI: 10.1016/j.taap.2008.12.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2008] [Revised: 11/21/2008] [Accepted: 12/02/2008] [Indexed: 11/24/2022]
Abstract
Nickel compounds are carcinogenic to humans and have been shown to alter epigenetic homeostasis. The c-Myc protein controls 15% of human genes and it has been shown that fluctuations of c-Myc protein alter global epigenetic marks. Therefore, the regulation of c-Myc by nickel ions in immortalized but not tumorigenic human bronchial epithelial Beas-2B cells was examined in this study. It was found that c-Myc protein expression was increased by nickel ions in non-tumorigenic Beas-2B and human keratinocyte HaCaT cells. The results also indicated that nickel ions induced apoptosis in Beas-2B cells. Knockout of c-Myc and its restoration in a rat cell system confirmed the essential role of c-Myc in nickel ion-induced apoptosis. Further studies in Beas-2B cells showed that nickel ion increased the c-Myc mRNA level and c-Myc promoter activity, but did not increase c-Myc mRNA and protein stability. Moreover, nickel ion upregulated c-Myc in Beas-2B cells through the MEK/ERK pathway. Collectively, the results demonstrate that c-Myc induction by nickel ions occurs via an ERK-dependent pathway and plays a crucial role in nickel-induced apoptosis in Beas-2B cells.
Collapse
Affiliation(s)
- Qin Li
- New York University School of Medicine, Nelson Institute of Environmental Medicine, 57 Old Forge Road, NY 10987, USA
| | - Ting-Chung Suen
- New York University School of Medicine, Nelson Institute of Environmental Medicine, 57 Old Forge Road, NY 10987, USA
| | - Hong Sun
- New York University School of Medicine, Nelson Institute of Environmental Medicine, 57 Old Forge Road, NY 10987, USA
| | - Adriana Arita
- New York University School of Medicine, Nelson Institute of Environmental Medicine, 57 Old Forge Road, NY 10987, USA
| | - Max Costa
- New York University School of Medicine, Nelson Institute of Environmental Medicine, 57 Old Forge Road, NY 10987, USA
| |
Collapse
|
46
|
Abstract
Although carcinogenic metals have been known to disrupt a wide range of cellular processes the precise mechanism by which these exert their carcinogenic effects is not known. Over the last decade or two, studies in the field of metal carcinogenesis suggest that epigenetic mechanisms may play a role in metal-induced carcinogenesis. In this review we summarize the evidence demonstrating that exposure to carcinogenic metals such as nickel, arsenic, chromium, and cadmium can perturb DNA methylation levels as well as global and gene specific histone tail posttranslational modification marks. We also wish to emphasize the importance in understanding that gene expression can be regulated by both genetic and epigenetic mechanisms and both these must be considered when studying the mechanism underlying the toxicity and cell-transforming ability of carcinogenic metals and other toxicants, and aberrant changes in gene expression that occur during disease states such as cancer.
Collapse
Affiliation(s)
- Adriana Arita
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 57 Old Forge Road, NY 10987, USA
| | - Max Costa
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 57 Old Forge Road, NY 10987, USA
| |
Collapse
|
47
|
Cummings SD, Ryu B, Samuels MA, Yu X, Meeker AK, Healey MA, Alani RM. Id1 delays senescence of primary human melanocytes. Mol Carcinog 2008; 47:653-9. [PMID: 18240291 DOI: 10.1002/mc.20422] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The Id family of helix-loop-helix transcription factors is upregulated in a variety of human malignancies and has been implicated in promoting tumorigenesis through effects on cell growth, differentiation, and tumor angiogenesis. While expression of Id proteins has been associated with tumorigenesis, the precise mechanistic relationship between Id expression and carcinogenesis has not been clearly delineated. We have previously shown that Id1 delays cellular senescence in primary mammalian cells through inhibition of the cell cycle regulatory protein and familial melanoma gene, p16/INK4a. We have also demonstrated that Id1 expression is upregulated in early stage primary human melanomas and may be an important marker for early malignancy. In order to further define the role of Id1 in human melanoma development, we have evaluated the function of Id1 in primary human melanocytes. Here we show that constitutive expression of Id1 in primary human melanocytes leads to delayed cellular senescence and decreased expression of the familial melanoma gene, p16/INK4a. Although melanocytes constitutively expressing Id1 are shown to possess extended lifespans, this is not associated with an appreciable change in cell growth or telomere length. We conclude that Id1 delays cellular senescence in primary human melanocytes through inhibition of p16/INK4a expression and suggest that Id1 may contribute to the malignant conversion of primary human melanocytes through extension of cellular lifespan.
Collapse
Affiliation(s)
- Staci D Cummings
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231-1000, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Reddy KK, Lefkove B, Chen LB, Govindarajan B, Carracedo A, Velasco G, Carrillo CO, Bhandarkar SS, Owens MJ, Mechta-Grigoriou F, Arbiser JL. The antidepressant sertraline downregulates Akt and has activity against melanoma cells. Pigment Cell Melanoma Res 2008; 21:451-6. [DOI: 10.1111/j.1755-148x.2008.00481.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
49
|
Ji W, Yang L, Yu L, Yuan J, Hu D, Zhang W, Yang J, Pang Y, Li W, Lu J, Fu J, Chen J, Lin Z, Chen W, Zhuang Z. Epigenetic silencing of O6-methylguanine DNA methyltransferase gene in NiS-transformed cells. Carcinogenesis 2008; 29:1267-75. [PMID: 18204074 DOI: 10.1093/carcin/bgn012] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2024] Open
Abstract
Nickel (Ni) compounds are potent carcinogens and can induce malignant transformation of rodent and human cells. To uncover the molecular mechanisms of nickel sulfide (NiS)-induced cell transformation, we investigated epigenetic alterations in a set of DNA repair genes. The silencing of the O(6)-methylguanine DNA methyltransferase (MGMT) gene locus and upregulation of DNA methyltransferase 1 (DNMT1) expression was specifically detected in NiS-transformed human bronchial epithelial (16HBE) cells. In addition, we noted epigenetic alterations including DNA hypermethylation, reduced histone H4 acetylation and a decrease in the ratio of Lys-9 acetylated/methylated histone H3 at the MGMT CpG island in NiS-transformed 16HBE cells. Meanwhile, we identified concurrent binding of methyl-CpG-binding protein 2, methylated DNA-binding domain protein 2 and DNMT1 to the CpG island of the MGMT promoter, demonstrating that these components collaborate to maintain MGMT methylation in NiS-transformed cells. Moreover, depletion of DNMT1 by introduction of a small hairpin RNA construct into NiS-transformed cells resulted in a 30% inhibition of cell proliferation and led to increased MGMT gene expression by reversion of the epigenetic modifications at the MGMT promoter region. MGMT suppression and hypermethylation at the CpG island of the MGMT promoter occurred 6 days after NiS treatment, indicating that epigenetic modifications of MGMT might be an early event in tumorigenesis. Taken together, these observations demonstrate that epigenetic silencing of MGMT is associated with DNA hypermethylation, histone modifications and DNMT1 upregulation, which contribute to NiS-induced malignant transformation.
Collapse
Affiliation(s)
- Weidong Ji
- Faculty of Preventive Medicine, School of Public Health, Sun Yat-Sen University, 74 Zhongshan Road 2, Guangzhou 510080, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
In melanoma, at least four major signaling abnormalities have been described. They include beta-catenin deregulation (mutation/mislocalization), p16 loss, MAP kinase activation, and Akt activation. In this review, we discuss the role of the fourth pathway, known as the reactive oxygen driven tumor. The role of reactive oxygen in tumorigenesis is likely to relate to virtually all forms of cancer, and lends itself to specific therapies. These include blockade of reactive oxygen, resulting in decreased activation of NF-kappaB, which should sensitize tumors to chemotherapy and radiation. The phenotype of the reactive oxygen driven tumor can be monitored using available markers already in use in most hospital laboratories.
Collapse
Affiliation(s)
- Levi Fried
- Department of Dermatology, Emory University School of Medicine, Atlanta, GA, USA
| | | |
Collapse
|