1
|
Meaza I, Williams AR, Wise SS, Lu H, Pierce JW. Carcinogenic Mechanisms of Hexavalent Chromium: From DNA Breaks to Chromosome Instability and Neoplastic Transformation. Curr Environ Health Rep 2024:10.1007/s40572-024-00460-9. [PMID: 39466546 DOI: 10.1007/s40572-024-00460-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 10/30/2024]
Abstract
PURPOSE OF REVIEW Hexavalent chromium [Cr(VI)] is a well-established human carcinogen, yet the mechanisms by which it leads to carcinogenic outcomes is still unclear. As a driving factor in its carcinogenic mechanism, Cr(VI) causes DNA double strand breaks and break-repair deficiency, leading to the development of chromosome instability. Therefore, the aim of this review is to discuss studies assessing Cr(VI)-induced DNA double strand breaks, chromosome damage and instability, and neoplastic transformation including cell culture, experimental animal, human pathology and epidemiology studies. RECENT FINDINGS Recent findings confirm Cr(VI) induces DNA double strand breaks, chromosome instability and neoplastic transformation in exposed cells, animals and humans, emphasizing these outcomes as key steps in the mechanism of Cr(VI) carcinogenesis. Moreover, recent findings suggest chromosome instability is a key phenotype in Cr(VI)-neoplastically transformed clones and is an inheritable and persistent phenotype in exposed cells, once more suggesting chromosome instability as central in the carcinogenic mechanism. Although limited, some studies have demonstrated DNA damage and epigenetic modulation are also key outcomes in biopsies from chromate workers that developed lung cancer. Additionally, we also summarized new studies showing Cr(VI) causes genotoxic and clastogenic effects in cells from wildlife, such as sea turtles, whales, and alligators. Overall, across the literature, it is clear that Cr(VI) causes neoplastic transformation and lung cancer. Many studies measured Cr(VI)-induced increases in DNA double strand breaks, the most lethal type of breaks clearly showing that Cr(VI) is genotoxic. Unrepaired or inaccurately repaired breaks lead to the development of chromosome instability, which is a common phenotype in Cr(VI) exposed cells, animals, and humans. Indeed, many studies show Cr(VI) induces both structural and numerical chromosome instability. Overall, the large body of literature strongly supports the conclusion that Cr(VI) causes DNA double strand breaks, inhibits DNA repair and chromosome instability, which are key to the development of Cr(VI)-induced cell transformation.
Collapse
Affiliation(s)
- Idoia Meaza
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston St, , Rm 1422, Louisville, KY, USA
| | - Aggie R Williams
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston St, , Rm 1422, Louisville, KY, USA
| | - Sandra S Wise
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston St, , Rm 1422, Louisville, KY, USA
| | - Haiyan Lu
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston St, , Rm 1422, Louisville, KY, USA
| | - John W Pierce
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston St, , Rm 1422, Louisville, KY, USA.
| |
Collapse
|
2
|
Fischer F, Stößer S, Wegmann L, Veh E, Lumpp T, Parsdorfer M, Schumacher P, Hartwig A. Chromate Affects Gene Expression and DNA Methylation in Long-Term In Vitro Experiments in A549 Cells. Int J Mol Sci 2024; 25:10129. [PMID: 39337613 PMCID: PMC11431867 DOI: 10.3390/ijms251810129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Chromate has been shown to dysregulate epigenetic mechanisms such as DNA methylation, leading to changes in gene expression and genomic instability. However, most in vitro studies are limited to short incubation periods, although chronic exposure may be more relevant for both environmental and occupational exposure. In this study, human adenocarcinoma A549 cells were treated with 1, 2 or 5 µM chromate for 24 h and compared with incubations with 0.2, 0.5 or 1 µM chromate for 1 to 5 weeks. Chromium accumulated in a pronounced time- and concentration-dependent manner after short-term treatment, whereas a plateau of intracellular chromium content was observed after long-term treatment. While short-term treatment induced a G2 arrest of the cell cycle, this effect was not observed after long-term treatment at lower concentrations. The opposite was observed for global DNA methylation: while short-term treatment showed no effect of chromate, significant dose-dependent hypomethylation was observed in the long-term experiments. Time-dependent effects were also observed in a high-throughput RT-qPCR gene expression analysis, particularly in genes related to the inflammatory response and DNA damage response. Taken together, the results suggest specific differences in toxicity profiles when comparing short-term and long-term exposure to chromate in A549 cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Andrea Hartwig
- Department of Food Chemistry and Toxicology, Institute of Applied Biosciences (IAB), Karlsruhe Institute of Technology (KIT), 76131 Karlsruhe, Germany
| |
Collapse
|
3
|
Lu H, Wise SS, Speer RM, Croom-Perez TJ, Toyoda JH, Meaza I, Williams A, Wise JP, Kouokam JC, Young Wise J, Hoyle GW, Zhu C, Ali AM, Wise JP. Acute particulate hexavalent chromium exposure induces DNA double-strand breaks and activates homologous recombination repair in rat lung tissue. Toxicol Sci 2024; 201:1-13. [PMID: 38867691 PMCID: PMC11347773 DOI: 10.1093/toxsci/kfae076] [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] [Indexed: 06/14/2024] Open
Abstract
Hexavalent chromium [Cr(VI)] is an established human lung carcinogen, but the carcinogenesis mechanism is poorly understood. Chromosome instability, a hallmark of lung cancer, is considered a major driver of Cr(VI)-induced lung cancer. Unrepaired DNA double-strand breaks are the underlying cause, and homologous recombination repair is the primary mechanism preventing Cr(VI)-induced DNA breaks from causing chromosome instability. Cell culture studies show acute Cr(VI) exposure causes DNA double-strand breaks and increases homologous recombination repair activity. However, the ability of Cr(VI)-induced DNA breaks and repair impact has only been reported in cell culture studies. Therefore, we investigated whether acute Cr(VI) exposure could induce breaks and homologous recombination repair in rat lungs. Male and female Wistar rats were acutely exposed to either zinc chromate particles in a saline solution or saline alone by oropharyngeal aspiration. This exposure route resulted in increased Cr levels in each lobe of the lung. We found Cr(VI) induced DNA double-strand breaks in a concentration-dependent manner, with females being more susceptible than males, and induced homologous recombination repair at similar levels in both sexes. Thus, these data show this driving mechanism discovered in cell culture indeed translates to lung tissue in vivo.
Collapse
Affiliation(s)
- Haiyan Lu
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
| | - Sandra S Wise
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
| | - Rachel M Speer
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
| | - Tayler J Croom-Perez
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
| | - Jennifer H Toyoda
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
| | - Idoia Meaza
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
| | - Aggie Williams
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
| | - John Pierce Wise
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
- Pediatric Research Institute, University of Louisville, Louisville, KY 40292, United States
| | - J Calvin Kouokam
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
| | - Jamie Young Wise
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
| | - Gary W Hoyle
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY 40292, United States
| | - Cairong Zhu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan 610044, China
| | - Abdul-Mehdi Ali
- Earth and Planetary Sciences Department, The University of New Mexico, Albuquerque, NM 87131, United States
| | - John Pierce Wise
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, United States
| |
Collapse
|
4
|
Salvatierra-Fréchou DM, Verstraeten SV. Tl(I) and Tl(III)-induce genotoxicity, reticulum stress and autophagy in PC12 Adh cells. Arch Toxicol 2024; 98:2085-2100. [PMID: 38619592 DOI: 10.1007/s00204-024-03752-z] [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: 02/09/2024] [Accepted: 03/21/2024] [Indexed: 04/16/2024]
Abstract
Thallium (Tl) and its two cationic species, Tl(I) and Tl(III), are toxic for most living beings. In this work, we investigated the effects of Tl (10-100 µM) on the viability and proliferation capacity of the adherent variant of PC12 cells (PC12 Adh cells). While both Tl(I) and Tl(III) halted cell proliferation from 24 h of incubation, their viability was ~ 90% even after 72 h of treatment. At 24 h, increased levels of γH2AX indicated the presence of DNA double-strand breaks. Simultaneously, increased expression of p53 and its phosphorylation at Ser15 were observed, which were associated with decreased levels of p-AKTSer473 and p-mTORSer2448. At 72 h, the presence of large cytoplasmic vacuoles together with increased autophagy predictor values suggested that Tl may induce autophagy in these cells. This hypothesis was corroborated by images obtained by transmission electron microscopy (TEM) and from the decreased expression at 72 h of incubation of SQSTM-1 and increased LC3β-II to LC3β-I ratio. TEM images also showed enlarged ER that, together with the increased expression of IRE1-α from 48 h of incubation, indicated that Tl-induced ER stress preceded autophagy. The inhibition of autophagy flux with chloroquine increased cell mortality, suggesting that autophagy played a cytoprotective role in Tl toxicity in these cells. Together, results indicate that Tl(I) or Tl(III) are genotoxic to PC12 Adh cells which respond to the cations inducing ER stress and cytoprotective autophagy.
Collapse
Affiliation(s)
- Damiana M Salvatierra-Fréchou
- Facultad de Farmacia y Bioquímica, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Universidad de Buenos Aires, Junín 956, C1113AAD, Buenos Aires, Argentina
| | - Sandra V Verstraeten
- Facultad de Farmacia y Bioquímica, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Universidad de Buenos Aires, Junín 956, C1113AAD, Buenos Aires, Argentina.
| |
Collapse
|
5
|
Tumolo M, De Paola D, Uricchio VF, Ancona V. Biostimulation effect of different amendments on Cr(VI) recovering microbial community. N Biotechnol 2023; 78:29-41. [PMID: 37793602 DOI: 10.1016/j.nbt.2023.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 09/15/2023] [Accepted: 09/30/2023] [Indexed: 10/06/2023]
Abstract
The present study used Cr(VI)-polluted microcosms amended with lactate or yeast extract, and nonamended microcosms as control, to investigate how a native bacterial community varied in response to the treatment and during the pollutant removal. Results suggested that providing electron donors resulted in a proliferation of a few bacterial species, with the consequent decrease in observed species richness and evenness, and was a driving force for the bacterial compositional shift. Lactate promoted, in the first instance, the enrichment of fermentative bacteria belonging to Chromobacteriaceae, including Paludibacterium, and Micrococcaceae as observed after 4 days. When the rate of Cr(VI) removal was maximum in microcosms amended with lactate, the most represented taxa were Pseudarcicella and Azospirillum. Using yeast extract as a carbon source and electron donor led instead to the significant enrichment of Shewanella, followed by Vogesella and Acinetobacter on the 4th day, corresponding to 90% of Cr(VI) removed from the system. After the complete Cr(VI) removal, achieved in 7 days in the presence of yeast extract, α-diversity was notably increased. The amendment-specific turnover of the enriched bacterial taxa resulted in a different kinetic of pollutant removal. In particular, yeast extract promoted the quickest Cr(VI) reduction, while lactate supported a slower, but also considerable, pollutant removal from water. Since it is reasonable to assume that a macroscopic effect, such as the observed Cr(VI) removal, involved the overrepresented taxa, deepening the knowledge of the native bacterial community and its changes were used to hypothesize the possible microbial pathways involved.
Collapse
Affiliation(s)
- Marina Tumolo
- Water Research Institute, Italian National Research Council (IRSA-CNR), 70132 Bari, BA, Italy; Department of Biology, University of Bari, 70126 Bari, BA, Italy
| | - Domenico De Paola
- Institute of Biosciences and Bioresources, Italian National Research Council (IBBR-CNR), 70126 Bari, BA, Italy.
| | - Vito Felice Uricchio
- Water Research Institute, Italian National Research Council (IRSA-CNR), 70132 Bari, BA, Italy
| | - Valeria Ancona
- Water Research Institute, Italian National Research Council (IRSA-CNR), 70132 Bari, BA, Italy.
| |
Collapse
|
6
|
Meaza I, Williams AR, Lu H, Kouokam JC, Toyoda JH, Croom-Perez TJ, Wise SS, Aboueissa AEM, Wise JP. Prolonged particulate hexavalent chromium exposure induces RAD51 foci inhibition and cytoplasmic accumulation in immortalized and primary human lung bronchial epithelial cells. Toxicol Appl Pharmacol 2023; 479:116711. [PMID: 37805091 PMCID: PMC10841504 DOI: 10.1016/j.taap.2023.116711] [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: 08/16/2023] [Revised: 10/04/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Hexavalent chromium [Cr(VI)] is a human lung carcinogen with widespread exposure risks. Cr(VI) causes DNA double strand breaks that if unrepaired, progress into chromosomal instability (CIN), a key driving outcome in Cr(VI)-induced tumors. The ability of Cr(VI) to cause DNA breaks and inhibit repair is poorly understood in human lung epithelial cells, which are extremely relevant since pathology data show Cr(VI)-induced tumors originate from bronchial epithelial cells. In the present study, we considered immortalized and primary human bronchial epithelial cells. Cells were treated with zinc chromate at concentrations ranging 0.05 to 0.4μg/cm2 for acute (24 h) and prolonged (120 h) exposures. DNA double strand breaks (DSBs) were measured by neutral comet assay and the status of homologous recombination repair, the main pathway to fix Cr(VI)-induced DSBs, was measured by RAD51 foci formation with immunofluorescence, RAD51 localization with confocal microscopy and sister chromatid exchanges. We found acute and prolonged Cr(VI) exposure induced DSBs. Acute exposure induced homologous recombination repair, but prolonged exposure inhibited it resulting in chromosome instability in immortalized and primary human bronchial epithelial cells.
Collapse
Affiliation(s)
- Idoia Meaza
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston Street, Building 55A, Room 1422, Louisville, KY 40292, United States of America
| | - Aggie R Williams
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston Street, Building 55A, Room 1422, Louisville, KY 40292, United States of America
| | - Haiyan Lu
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston Street, Building 55A, Room 1422, Louisville, KY 40292, United States of America
| | - J Calvin Kouokam
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston Street, Building 55A, Room 1422, Louisville, KY 40292, United States of America
| | - Jennifer H Toyoda
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston Street, Building 55A, Room 1422, Louisville, KY 40292, United States of America
| | - Tayler J Croom-Perez
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 6900 Lake Nona Blvd., Orlando, FL 32827, United States of America
| | - Sandra S Wise
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston Street, Building 55A, Room 1422, Louisville, KY 40292, United States of America
| | | | - John Pierce Wise
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston Street, Building 55A, Room 1422, Louisville, KY 40292, United States of America.
| |
Collapse
|
7
|
Valiente S, Krawic C, Zhitkovich A. ATR activation by Cr-DNA damage is a major survival response establishing late S and G2 checkpoints after Cr(VI) exposure. Toxicol Appl Pharmacol 2023; 477:116696. [PMID: 37734571 PMCID: PMC10591798 DOI: 10.1016/j.taap.2023.116696] [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: 08/03/2023] [Revised: 09/17/2023] [Accepted: 09/18/2023] [Indexed: 09/23/2023]
Abstract
Inhalation exposure to hexavalent chromium is known to cause lung cancer and other pulmonary toxicity. Cellular metabolism of chromium(VI) entering cells as chromate anion produces different amounts of reactive Cr(V) intermediates and finally yields Cr(III). Direct reduction of Cr(VI) by ascorbate (Asc), the dominant metabolic reaction in vivo but not in standard cell cultures, skips production of Cr(V) but still permits extensive formation of Cr-DNA damage. To understand the importance of different forms of biological injury in Cr(VI) toxicity, we examined activation of several protein- and DNA damage-sensitive stress responses in human lung cells under Asc-restored conditions. We found that Asc-restored cells suppressed upregulation of oxidant-sensitive stress systems by Cr(VI) but showed a strong activation of the apical DNA damage-responsive kinase ATR. ATR signaling was triggered in late S phase and persisted upon entry of cells into G2 phase. Inhibition of ATR prevented the establishment of late-S and G2 cell cycle checkpoints and did not lead to a compensatory activation of a related kinase ATM. Inactivation of ATR also strongly impaired viability of Cr(VI)-treated lung cells including stem-like cells and revealed a significant formation of toxic Cr-DNA damage at low Cr(VI) doses. Our findings identified a major Cr(VI) resistance mechanism involving sensing of Cr-DNA damage by ATR in late S phase and a subsequent establishment of protective cell cycle checkpoints.
Collapse
Affiliation(s)
- Sophia Valiente
- Brown University, Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Providence, RI 02912, USA
| | - Casey Krawic
- Brown University, Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Providence, RI 02912, USA
| | - Anatoly Zhitkovich
- Brown University, Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Providence, RI 02912, USA.
| |
Collapse
|
8
|
Zhuo G, Wang L, Ali M, Jing Z, Hassan MF. Effect of hexavalent chromium on growth performance and metabolism in broiler chicken. Front Vet Sci 2023; 10:1273944. [PMID: 37822955 PMCID: PMC10562699 DOI: 10.3389/fvets.2023.1273944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/07/2023] [Indexed: 10/13/2023] Open
Abstract
Hexavalent chromium Cr (VI) is one of the most hazardous heavy metals in the environment and is toxic to living organisms causing tissue damage, disruption of the intestinal microbiota and cancer. However, there is little information on the relationship between the Cr (VI) and broiler chickens. The current study was performed to investigate the effect of Cr (VI) on growth performance, serum biochemical analysis, histopathological observations, and metabolomics analysis in broilers. Results show that Cr (VI) exposure significantly decreased the body weight (p < 0.01) and caused liver damages in broilers. With the extension of Cr (VI) action time, the liver appeared obvious pathological changes, including hepatic cord disorder, incomplete hepatocyte additionally, decreased serum biochemical indices of calcium (Ca), phosphorus (P), total protein (TP), phosphatase (ALP), and globin (GLB) significantly (p < 0.01). Moreover, metabolomics analysis indicated that 29 differential metabolites were identified, such as phytosphingosine, L-Serine, 12, 13-DHOME, Alpha-dimorphecolic acid, L-Methionine, L-Phenylalanine, 3-Dehydroshikimate, L-Tyrosine, and N-Acetyl-L-phenylalanine were significantly decreased under the action of Cr (VI) (p < 0.05). These 29 differential metabolites are mainly involved in 35 metabolic pathways, such as aminoacyl-tRNA biosynthesis, phenylalanine metabolism, sphingolipid, and linoleic metabolism. The study revealed that exposure to Cr (VI) resulted in a decrease in growth performance and metabolism, with the hazards and toxicity in broiler chicken. The findings provided new insight and a comprehensive understanding of the relationship between Cr (VI) and broiler chickens.
Collapse
Affiliation(s)
- Guorong Zhuo
- College of Small Animal Science and Technology, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, China
| | - Lei Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Muhammad Ali
- Basic Health Unit, Department of Health, Dera Ghazi Khan, Pakistan
| | - Zheng Jing
- College of Small Animal Science and Technology, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, China
| | | |
Collapse
|
9
|
Mezencev R, Gibbons C. Interactions between chromium species and DNA in vitro and their potential role in the toxicity of hexavalent chromium. Metallomics 2023; 15:mfad045. [PMID: 37491700 DOI: 10.1093/mtomcs/mfad045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/12/2023] [Indexed: 07/27/2023]
Abstract
Epidemiological and animal studies have supported the carcinogenicity of hexavalent chromium [Cr(VI)]; however, molecular changes responsible for the induction of cancer by Cr(VI) are not entirely understood. Numerous mechanistic studies suggested the role of oxidative stress and genotoxicity in Cr(VI)-mediated carcinogenesis; however, specific types of DNA damage have not yet been conclusively attributed to specific chromium species or other reactive byproducts generated in biological systems exposed to Cr(VI). Due to the remarkably complex chemistry and biological effects of chromium species generated through the intracellular reduction of Cr(VI), their relevance for Cr(VI)-mediated carcinogenesis has not yet been fully elucidated and continues to be a subject of ongoing discussions in the field. In this report, we describe a complex world of chromium species and their reactivity with DNA and other biologically relevant molecules in vitro to inform a more complete understanding of Cr(VI)-mediated toxicity. In addition, we discuss previous results in the context of in vitro models and analytical methods to reconcile some conflicting findings on the biological role of chromium species.
Collapse
Affiliation(s)
- R Mezencev
- Center for Public Health and Environmental Assessment, Office of Research and Development, US EPA, Washington, DC, USA
| | - C Gibbons
- Center for Public Health and Environmental Assessment, Office of Research and Development, US EPA, Washington, DC, USA
| |
Collapse
|
10
|
Zablon HA, VonHandorf A, Puga A. Mechanisms of chromate carcinogenesis by chromatin alterations. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 96:1-23. [PMID: 36858770 DOI: 10.1016/bs.apha.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In a dynamic environment, organisms must constantly mount an adaptive response to new environmental conditions in order to survive. Novel patterns of gene expression, driven by attendant changes in chromatin architecture, aid in adaptation and survival. Critical mechanisms in the control of gene transcription govern new spatiotemporal chromatin-chromatin interactions that make regulatory DNA elements accessible to the transcription factors that control the response. Consequently, agents that disrupt chromatin structure are likely to have a direct impact on the transcriptional programs of cells and organisms and to drive alterations in fundamental physiological processes. In this regard, hexavalent chromium (Cr(VI)) is of special interest because it interacts directly with cellular proteins, DNA, and other macromolecules, and is likely to upset cell functions that may cause generalized damage to the organism. Here, we will highlight chromium-mediated mechanisms that disrupt chromatin architecture and discuss how these mechanisms are integral to its carcinogenic properties. Emerging evidence indicates that Cr(VI) targets euchromatin, particularly in genomic locations flanking the binding sites of the essential transcription factors CTCF and AP1, and, in so doing, they disrupt nucleosomal architecture. Ultimately, the ensuing changes, if occurring in critical regulatory domains, may establish a new chromatin state, either toxic or adaptive, that will be governed by the corresponding gene transcription changes in key biological processes associated with that state.
Collapse
Affiliation(s)
- Hesbon A Zablon
- Department of Environmental and Public Health Sciences and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Andrew VonHandorf
- Department of Environmental and Public Health Sciences and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Alvaro Puga
- Department of Environmental and Public Health Sciences and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.
| |
Collapse
|
11
|
Batyrova G, Kononets V, Amanzholkyzy A, Tlegenova Z, Umarova G. Chromium as a Risk Factor for Breast Cancer: A Meta-Analysis. Asian Pac J Cancer Prev 2022; 23:3993-4003. [PMID: 36579979 PMCID: PMC9971475 DOI: 10.31557/apjcp.2022.23.12.3993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Chromium (Cr) is a transition metal, natural element. Chromium is the 21st most abundant element in Earth's crust. Cr is found in soil, rocks and living organisms. It may have various oxidation states, from -2 to +6, but most of these states are too unstable to exist in any significant quantities. The purpose of this review and meta-analysis is to critically assess the scientific evidence on the carcinogenic effects of chromium (Cr) and to determine whether there is currently sufficient evidence to suggest that that there is a link between chromium levels in hair and blood serum and breast cancer in women. MATERIAL AND METHODS Research on the relationship between heavy metal chromium and the risk of developing breast cancer has been searched in PubMed, EMBASE, Web of Science, Scopus among papers published between January 2000 and September 2020. The search used the following terms (MeSH): breast cancer, women, trace elements, metals, chromium, chemically-induced, hair, serum using additional terms. RESULTS In the second group of comparisons of women from "ecologically clean" districts of Aktobe Region, there were significantly lower indicators of the microelements in tumor tissue. The amount of Fe ranges from 38.46 to 65.39 ug/g (average 49.56±5.81 ug/g), Cu from 2.8 to 6.69 ug/g (average 5.06±1.01 ug/g), Zn from 1.89 to 5.38 ug/g (average 3.88±0.89 ug/g), Cr from zero to 6,1 ug/g (average 2.13±1.29 ug/g), Ni from 0.11 to 0.42 ug/g (average 0.28±0.067 ug/g) и Pb from zero to 0.19 ug/g (average 0.098±0.06 ug/g). CONCLUSION The article established that women who live or work in ecologically polluted areas or have problems with micronutrient exchange need in-depth screening and more frequent screening for early detection of pre- and breast cancer.
Collapse
Affiliation(s)
- Gulnara Batyrova
- Department of Clinical Laboratory and Visual Diagnostics, West Kazakhstan Marat Ospanov Medical University, Aktobe, Republic of Kazakhstan. ,For Correspondence:
| | - Victoria Kononets
- Department of Molecular Biology and Medical Genetics, West Kazakhstan Marat Ospanov Medical University, Aktobe, Republic of Kazakhstan.
| | - Ainur Amanzholkyzy
- Department of Normal Phiziology, West Kazakhstan Marat Ospanov Medical University, Aktobe, Republic of Kazakhstan.
| | - Zhenisgul Tlegenova
- Department of Internal Diseases No. 2, West Kazakhstan Marat Ospanov Medical University, Aktobe, Republic of Kazakhstan.
| | - Gulmira Umarova
- Department of Evidence-Based Medicine and Scientific Management, West Kazakhstan Marat Ospanov Medical University, Aktobe, Republic of Kazakhstan.
| |
Collapse
|
12
|
Krawic C, Zhitkovich A. Chemical mechanisms of DNA damage by carcinogenic chromium(VI). ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 96:25-46. [PMID: 36858775 PMCID: PMC10069994 DOI: 10.1016/bs.apha.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hexavalent chromium is a firmly established human carcinogen with documented exposures in many professional groups. Environmental exposure to Cr(VI) is also a significant public health concern. Cr(VI) exists in aqueous solutions as chromate anion that is unreactive with DNA and requires reductive activation inside the cells to produce genotoxic and mutagenic effects. Reduction of Cr(VI) in cells is nonenzymatic and in vivo principally driven by ascorbate with a secondary contribution from nonprotein thiols glutathione and cysteine. In addition to its much faster rate of reduction, ascorbate-driven metabolism avoids the formation of Cr(V) which is the first intermediate in Cr(VI) reduction by thiols. The end-product of Cr(VI) reduction is Cr(III) which forms several types of Cr-DNA adducts that are collectively responsible for all mutagenic and genotoxic effects in Cr(VI) reactions with ascorbate and thiols. Some Cr(V) forms can react with H2O2 to produce DNA-oxidizing peroxo species although this genotoxic pathway is suppressed in cells with physiological levels of ascorbate. Chemical reactions of Cr(VI) with ascorbate or thiols lack directly DNA-oxidizing metabolites. The formation of oxidative DNA breaks in early studies of these reactions was caused by iron contamination. Production of Cr(III)-DNA adducts in cells showed linear dose-dependence irrespective of the predominant reduction pathway and their processing by mismatch repair generated more toxic secondary genetic lesions in euchromatin. Overall, Cr(III)-DNA adduction is the dominant pathway for the formation of genotoxic and mutagenic DNA damage by carcinogenic Cr(VI).
Collapse
Affiliation(s)
- Casey Krawic
- Department of Pathology and Laboratory Medicine, Legorreta Cancer Center, Brown University, Providence, RI, United States
| | - Anatoly Zhitkovich
- Department of Pathology and Laboratory Medicine, Legorreta Cancer Center, Brown University, Providence, RI, United States.
| |
Collapse
|
13
|
Unveiling the Toxicity of Fine and Nano-Sized Airborne Particles Generated from Industrial Thermal Spraying Processes in Human Alveolar Epithelial Cells. Int J Mol Sci 2022; 23:ijms23084278. [PMID: 35457096 PMCID: PMC9025379 DOI: 10.3390/ijms23084278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/09/2022] [Accepted: 04/10/2022] [Indexed: 02/04/2023] Open
Abstract
High-energy industrial processes have been associated with particle release into workplace air that can adversely affect workers’ health. The present study assessed the toxicity of incidental fine (PGFP) and nanoparticles (PGNP) emitted from atmospheric plasma (APS) and high-velocity oxy-fuel (HVOF) thermal spraying. Lactate dehydrogenase (LDH) release, 2-(4-nitrophenyl)-2H-5-tetrazolio]-1,3-benzene disulfonate (WST-1) metabolisation, intracellular reactive oxygen species (ROS) levels, cell cycle changes, histone H2AX phosphorylation (γ-H2AX) and DNA damage were evaluated in human alveolar epithelial cells at 24 h after exposure. Overall, HVOF particles were the most cytotoxic to human alveolar cells, with cell viability half-maximal inhibitory concentration (IC50) values of 20.18 µg/cm2 and 1.79 µg/cm2 for PGFP and PGNP, respectively. Only the highest tested concentration of APS-PGFP caused a slight decrease in cell viability. Particle uptake, cell cycle arrest at S + G2/M and γ-H2AX augmentation were observed after exposure to all tested particles. However, higher levels of γ-H2AX were found in cells exposed to APS-derived particles (~16%), while cells exposed to HVOF particles exhibited increased levels of oxidative damage (~17% tail intensity) and ROS (~184%). Accordingly, APS and HVOF particles seem to exert their genotoxic effects by different mechanisms, highlighting that the health risks of these process-generated particles at industrial settings should not be underestimated.
Collapse
|
14
|
Zhou Z, Yin H, Suye S, Zhu F, Cai H, Fu C. The changes of DNA double-strand breaks and DNA repair during ovarian reserve formation in mice. Reprod Biol 2022; 22:100603. [PMID: 35026551 DOI: 10.1016/j.repbio.2022.100603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 12/16/2021] [Accepted: 01/02/2022] [Indexed: 11/19/2022]
Abstract
DNA double-strand break (DSB) repair is crucial to maintain genomic stability for sufficient ovarian reserve. It remains unknown the changes of DSBs formation and DNA repair in germ cells during ovarian reserve formation in FVB/N mice. We demonstrated germ cell numbers increased significantly (all P < 0.05) from E11.5 to E13.5 and decreased significantly (all P> 0.05) until P2. OCT4 and SOX2 analyses indicated pluripotency peaks at E13.5 then decreases significantly (all P 0.05) until P2. γH2AX analyses revealed DSB formation significantly (P < 0.05) increased from E13.5 until P2. RAD51 and DMC1 data revealed homologous recombination (HR) pathway repair of DSBs is persistent active during meiosis (E13.5- P2) (all P> 0.05). 53BP1 and KU70 data indicate the non-homologous end-joining pathway (NHEJ) remains active during meiosis. 53BP1 expression was highest at E13.5 (P < 0.05). KU70 expression was higher in germ cells from E15.5 to P2 (all < P 0.05). PH3 and KI67 analyses revealed germ cell proliferation was not significantly different (all P> 0.05) from E13.5 to P2. Caspase-3 and TUNEL analyses showed germ cells apoptosis was not significantly different (all P > 0.05) from E13.5 to P2. In conclusion, we found both germ cell number and pluripotency peak at E13.5 and decline during meiosis. We demonstrated HR and NHEJ continually repair DSBs during meiosis. RAD51 and DMC1 are continuously expressed during meiosis. 53BP1 is mainly expressed at E13.5. KU70 continually functions from E15.5 to P2. Proliferating and apoptotic cells were rarely detected during meiosis. Results provide a basis for further study of how DSBs and DNA repair affect germ cell development.
Collapse
Affiliation(s)
- Zhixian Zhou
- Department of Obstetrics and Gynecology, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Huan Yin
- Department of Obstetrics and Gynecology, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Suye Suye
- Department of Obstetrics and Gynecology, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Fang Zhu
- Department of Obstetrics and Gynecology, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Haiyi Cai
- Department of Clinical Medicine, Harbin Medical University, Harbin, 150081, China
| | - Chun Fu
- Department of Obstetrics and Gynecology, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
15
|
VonHandorf A, Zablon HA, Puga A. Hexavalent chromium disrupts chromatin architecture. Semin Cancer Biol 2021; 76:54-60. [PMID: 34274487 PMCID: PMC8627925 DOI: 10.1016/j.semcancer.2021.07.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 12/21/2022]
Abstract
Accessibility of DNA elements and the orchestration of spatiotemporal chromatin-chromatin interactions are critical mechanisms in the regulation of gene transcription. Thus, in an ever-changing milieu, cells mount an adaptive response to environmental stimuli by modulating gene expression that is orchestrated by coordinated changes in chromatin architecture. Correspondingly, agents that alter chromatin structure directly impact transcriptional programs in cells. Heavy metals, including hexavalent chromium (Cr(VI)), are of special interest because of their ability to interact directly with cellular protein, DNA and other macromolecules, resulting in general damage or altered function. In this review we highlight the chromium-mediated mechanisms that promote disruption of chromatin architecture and how these processes are integral to its carcinogenic properties. Emerging evidence shows that Cr(VI) targets nucleosomal architecture in euchromatin, particularly in genomic locations flanking binding sites of the essential transcription factors CTCF and AP1. Ultimately, these changes contribute to an altered chromatin state in critical gene regulatory regions, which disrupts gene transcription in functionally relevant biological processes.
Collapse
Affiliation(s)
- Andrew VonHandorf
- Department of Environmental and Public Health Sciences and Center for Environmental Genetics, University of Cincinnati College of Medicine, 160 Panzeca Way, Cincinnati, OH, 45267, USA
| | - Hesbon A Zablon
- Department of Environmental and Public Health Sciences and Center for Environmental Genetics, University of Cincinnati College of Medicine, 160 Panzeca Way, Cincinnati, OH, 45267, USA
| | - Alvaro Puga
- Department of Environmental and Public Health Sciences and Center for Environmental Genetics, University of Cincinnati College of Medicine, 160 Panzeca Way, Cincinnati, OH, 45267, USA.
| |
Collapse
|
16
|
DNA Double-Strand Breaks Induced in Human Cells by Twelve Metallic Species: Quantitative Inter-Comparisons and Influence of the ATM Protein. Biomolecules 2021; 11:biom11101462. [PMID: 34680095 PMCID: PMC8533583 DOI: 10.3390/biom11101462] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/29/2021] [Accepted: 10/02/2021] [Indexed: 01/25/2023] Open
Abstract
Despite a considerable amount of data, the molecular and cellular bases of the toxicity due to metal exposure remain unknown. Recent mechanistic models from radiobiology have emerged, pointing out that the radiation-induced nucleo-shuttling of the ATM protein (RIANS) initiates the recognition and the repair of DNA double-strand breaks (DSB) and the final response to genotoxic stress. In order to document the role of ATM-dependent DSB repair and signalling after metal exposure, we applied twelve different metal species representing nine elements (Al, Cu, Zn Ni, Pd, Cd, Pb, Cr, and Fe) to human skin, mammary, and brain cells. Our findings suggest that metals may directly or indirectly induce DSB at a rate that depends on the metal properties and concentration, and tissue type. At specific metal concentration ranges, the nucleo-shuttling of ATM can be delayed which impairs DSB recognition and repair and contributes to toxicity and carcinogenicity. Interestingly, as observed after low doses of ionizing radiation, some phenomena equivalent to the biological response observed at high metal concentrations may occur at lower concentrations. A general mechanistic model of the biological response to metal exposure based on the nucleo-shuttling of ATM is proposed to describe the metal-induced stress response and to define quantitative endpoints for toxicity and carcinogenicity.
Collapse
|
17
|
Zhitkovich A. Ascorbate: antioxidant and biochemical activities and their importance for in vitro models. Arch Toxicol 2021; 95:3623-3631. [PMID: 34596731 DOI: 10.1007/s00204-021-03167-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/16/2021] [Indexed: 12/19/2022]
Abstract
Ascorbate has many biological activities that involve fundamental cellular functions such as gene expression, differentiation, and redox homeostasis. Biochemically, it serves as a cofactor for a large family of dioxygenases (> 60 members) which control transcription, formation of extracellular matrix, and epigenetic processes of histone and DNA demethylation. Ascorbate is also a major antioxidant acting as a very effective scavenger of primary reactive oxygen species. Reduction of Fe(III) by ascorbate is important for cellular uptake of iron via DMT1. Cell culture models are extensively used in toxicology and pharmacology for mechanistic studies of nutrients, drugs and other xenobiotics. High-throughput screens in vitro, such as a large-scale Tox21 program in the US, offers opportunities to assess hazardous properties of a vast and growing number of industrial chemicals. However, cells in typical cultures are severely deficient in ascorbate, raising concerns about their ability to accurately recapitulate toxic and other responses in vivo. Scarcity of ascorbate and a frequently unrecognized use of media with its thiol substitute alters stress sensitivity of cells in different directions. Remediation of ascorbate deficiency in tissue culture restores the physiological state of many cellular processes and it should improve a currently limited toxicity predictability of in vitro bioassays.
Collapse
Affiliation(s)
- Anatoly Zhitkovich
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship Street, Room 507, Providence, RI, 02912, USA.
| |
Collapse
|
18
|
Ventura C, Gomes BC, Oberemm A, Louro H, Huuskonen P, Mustieles V, Fernández MF, Ndaw S, Mengelers M, Luijten M, Gundacker C, Silva MJ. Biomarkers of effect as determined in human biomonitoring studies on hexavalent chromium and cadmium in the period 2008-2020. ENVIRONMENTAL RESEARCH 2021; 197:110998. [PMID: 33713715 DOI: 10.1016/j.envres.2021.110998] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 06/12/2023]
Abstract
A number of human biomonitoring (HBM) studies have presented data on exposure to hexavalent chromium [Cr(VI)] and cadmium (Cd), but comparatively few include results on effect biomarkers. The latter are needed to identify associations between exposure and adverse outcomes (AOs) in order to assess public health implications. To support improved derivation of EU regulation and policy making, it is of great importance to identify the most reliable effect biomarkers for these heavy metals that can be used in HBM studies. In the framework of the Human Biomonitoring for Europe (HBM4EU) initiative, our study aim was to identify effect biomarkers linking Cr(VI) and Cd exposure to selected AOs including cancer, immunotoxicity, oxidative stress, and omics/epigenetics. A comprehensive PubMed search identified recent HBM studies, in which effect biomarkers were examined. Validity and applicability of the markers in HBM studies are discussed. The most frequently analysed effect biomarkers regarding Cr(VI) exposure and its association with cancer were those indicating oxidative stress (e.g., 8-hydroxy-2'-deoxyguanosine (8-OHdG), malondialdehyde (MDA), glutathione (GSH)) and DNA or chromosomal damage (comet and micronucleus assays). With respect to Cd and to some extent Cr, β-2-microglobulin (B2-MG) and N-acetyl-β-D-glucosaminidase (NAG) are well-established, sensitive, and the most common effect biomarkers to relate Cd or Cr exposure to renal tubular dysfunction. Neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule (KIM)-1 could serve as sensitive biomarkers of acute kidney injury in response to both metals, but need further investigation in HBM studies. Omics-based biomarkers, i.e., changes in the (epi-)genome, transcriptome, proteome, and metabolome associated with Cr and/or Cd exposure, are promising effect biomarkers, but more HBM data are needed to confirm their significance. The combination of established effect markers and omics biomarkers may represent the strongest approach, especially if based on knowledge of mechanistic principles. To this aim, also mechanistic data were collected to provide guidance on the use of more sensitive and specific effect biomarkers. This also led to the identification of knowledge gaps relevant to the direction of future research.
Collapse
Affiliation(s)
- Célia Ventura
- National Institute of Health Doutor Ricardo Jorge (INSA), Human Genetics Department, Av. Padre Cruz, 1649-016, Lisbon, Toxicogenomics and Human Health (ToxOmics), NOVA Medical School/FCM, Universidade Nova de Lisboa, Portugal
| | - Bruno Costa Gomes
- National Institute of Health Doutor Ricardo Jorge (INSA), Human Genetics Department, Av. Padre Cruz, 1649-016, Lisbon, Toxicogenomics and Human Health (ToxOmics), NOVA Medical School/FCM, Universidade Nova de Lisboa, Portugal
| | - Axel Oberemm
- German Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Henriqueta Louro
- National Institute of Health Doutor Ricardo Jorge (INSA), Human Genetics Department, Av. Padre Cruz, 1649-016, Lisbon, Toxicogenomics and Human Health (ToxOmics), NOVA Medical School/FCM, Universidade Nova de Lisboa, Portugal
| | - Pasi Huuskonen
- Finnish Institute of Occupational Health, PO Box 40, FI-00032 Työterveyslaitos, Finland
| | - Vicente Mustieles
- Center for Biomedical Research (CIBM), University of Granada, Granada, Spain; Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, Spain; Consortium for Biomedical Research in Epidemiology & Public Health (CIBERESP), Spain
| | - Mariana F Fernández
- Center for Biomedical Research (CIBM), University of Granada, Granada, Spain; Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, Spain; Consortium for Biomedical Research in Epidemiology & Public Health (CIBERESP), Spain
| | - Sophie Ndaw
- French National Research and Safety Institute (INRS), France
| | - Marcel Mengelers
- National Institute for Public Health and the Environment (RIVM), Centre for Nutrition, Prevention and Health Services, Department of Food Safety, Bilthoven, the Netherlands
| | - Mirjam Luijten
- National Institute for Public Health and the Environment (RIVM), Centre for Health Protection, Bilthoven, the Netherlands
| | - Claudia Gundacker
- Institute of Medical Genetics, Medical University of Vienna, Waehringer Strasse 10, A-1090 Vienna, Austria.
| | - Maria João Silva
- National Institute of Health Doutor Ricardo Jorge (INSA), Human Genetics Department, Av. Padre Cruz, 1649-016, Lisbon, Toxicogenomics and Human Health (ToxOmics), NOVA Medical School/FCM, Universidade Nova de Lisboa, Portugal.
| |
Collapse
|
19
|
VonHandorf A, Zablon HA, Biesiada J, Zhang X, Medvedovic M, Puga A. Hexavalent chromium promotes differential binding of CTCF to its cognate sites in Euchromatin. Epigenetics 2021; 16:1361-1376. [PMID: 33319643 DOI: 10.1080/15592294.2020.1864168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Hexavalent chromium compounds are well-established respiratory carcinogens to which humans are commonly exposed in industrial and occupational settings. In addition, natural and anthropogenic sources of these compounds contribute to the exposure of global populations through multiple routes, including dermal, ingestion and inhalation that elevate the risk of cancer by largely unresolved mechanisms. Cr(VI) has genotoxic properties that include ternary adduct formation with DNA, increases in DNA damage, mostly by double-strand break formation, and altered transcriptional responses. Our previous work using ATAC-seq showed that CTCF motifs were enriched in Cr(VI)-dependent differentially accessible chromatin, suggesting that CTCF, a key transcription factor responsible for the regulation of the transcriptome, might be a chromium target. To test this hypothesis, we investigated the effect of Cr(VI) treatment on the binding of CTCF to its cognate sites and ensuing changes in transcription-related histone modifications. Differentially bound CTCF sites were enriched by Cr(VI) treatment within transcription-related regions, specifically transcription start sites and upstream genic regions. Functional annotation of the affected genes highlighted biological processes previously associated with Cr(VI) exposure. Notably, we found that differentially bound CTCF sites proximal to the promoters of this subset of genes were frequently associated with the active histone marks H3K27ac, H3K4me3, and H3K36me3, in agreement with the concept that Cr(VI) targets CTCF in euchromatic regions of the genome. Our results support the conclusion that Cr(VI) treatment promotes the differential binding of CTCF to its cognate sites in genes near transcription-active boundaries, targeting these genes for dysregulation.
Collapse
Affiliation(s)
- Andrew VonHandorf
- Department of Environmental and Public Health Sciences and Center for Environmental Genetics University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Hesbon A Zablon
- Department of Environmental and Public Health Sciences and Center for Environmental Genetics University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jacek Biesiada
- Department of Environmental and Public Health Sciences and Center for Environmental Genetics University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Xiang Zhang
- Department of Environmental and Public Health Sciences and Center for Environmental Genetics University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Mario Medvedovic
- Department of Environmental and Public Health Sciences and Center for Environmental Genetics University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Alvaro Puga
- Department of Environmental and Public Health Sciences and Center for Environmental Genetics University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
20
|
Abstract
![]()
Vitamin
C (ascorbic acid) is a water-soluble antioxidant and a
cofactor for a large number of enzymes. It is present in all tissues
and especially abundant in corneal epithelium, stem cells, and neurons.
Although similar to thiols in its ability to react with many reactive
oxygen species (ROS), ascorbate is much better (>100× faster)
than glutathione at scavenging of primary ROS (superoxide radical
and singlet oxygen). Ascorbate appears to be especially important
for elimination of O2•– in the
nucleus which contains little or no SOD activity. Cofactor functions
of ascorbate involve the maintenance of activity of Fe(II)/2-oxoglutarate-dependent
dioxygenases via reduction of Fe(III). The most prominent activity
of ascorbate-dependent dioxygenases in the cytoplasm is hydroxylation
of prolines in proteins involved in the formation of extracellular
matrix and regulation of metabolism and hypoxia responses. In the
nucleus, ascorbate is important for oxidative demethylation of 5-methylcytosine
in DNA (by TET proteins) and removal of methyl groups from histone
lysines (by JmjC demethylases). Differentiation and other cellular
reprograming processes involving DNA demethylation are especially
sensitive to ascorbate insufficiency. High doses of vitamin C alone
or in combinations with drugs produced cancer-suppressive effects
which involved redox, immune, and epigenetic mechanisms. Solutions
to vitamin C deficiency in cultured cells are discussed to improve
the physiological relevance of in vitro models. An
abundance of vitamin C in rodents limits their ability to fully recapitulate
human sensitivity to adverse health effects of malnutrition and xenobiotics,
including neurotoxicity, lung injury, and intergenerational and other
epigenetic effects.
Collapse
Affiliation(s)
- Anatoly Zhitkovich
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship Street, Providence, Rhode Island 02912, United States
| |
Collapse
|
21
|
Ruiz-Aguilar B, Raya-González J, López-Bucio JS, Reyes de la Cruz H, Herrera-Estrella L, Ruiz-Herrera LF, Martínez-Trujillo M, López-Bucio J. Mutation of MEDIATOR 18 and chromate trigger twinning of the primary root meristem in Arabidopsis. PLANT, CELL & ENVIRONMENT 2020; 43:1989-1999. [PMID: 32400913 DOI: 10.1111/pce.13786] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/25/2020] [Accepted: 05/06/2020] [Indexed: 06/11/2023]
Abstract
Plants adapt to soil injury and biotic stress via cell regeneration. In Arabidopsis, root tip damage by genotoxic agents, antibiotics, UV light and cutting induces a program that recovers the missing tissues through activation of stem cells and involves ethylene response factor 115 (ERF115), which triggers cell replenishment. Here, we show that mutation of the gene encoding an MED18 subunit of the transcriptional MEDIATOR complex and chromate [Cr(VI)], an environmental pollutant, synergistically trigger a developmental program that enables the splitting of the meristem in vivo to produce twin roots. Expression of the quiescent centre gene marker WOX5, auxin-inducible DR5:GFP reporter and the ERF115 factor traced the changes in cell identity during the conversion of single primary root meristems into twin roots and were induced in an MED18 and chromate-dependent manner during the root twinning events, which also required auxin redistribution and signalling mediated by IAA14/SOLITARY ROOT (SLR1). Splitting of the root meristem allowed dichotomous root branching in Arabidopsis, a poorly understood process in which stem cells may act to enable whole organ regeneration.
Collapse
Affiliation(s)
- Bricia Ruiz-Aguilar
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Javier Raya-González
- Facultad de Químico Farmacobiología, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Jesús Salvador López-Bucio
- CONACYT, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo. Edificio B3, Ciudad Universitaria, Morelia, Mexico
| | - Homero Reyes de la Cruz
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Luis Herrera-Estrella
- Unidad de Genómica Avanzada, Laboratorio Nacional de Genómica para la Biodiversidad, Centro de Investigación y de Estudios Avanzados del IPN, Campus Irapuato, Guanajuato, Mexico
| | - León Francisco Ruiz-Herrera
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Miguel Martínez-Trujillo
- Facultad de Biología, Universidad Michoacana de San Nicolás de Hidalgo. Edificio R, Ciudad Universitaria, Morelia, Mexico
| | - José López-Bucio
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| |
Collapse
|
22
|
Tumolo M, Ancona V, De Paola D, Losacco D, Campanale C, Massarelli C, Uricchio VF. Chromium Pollution in European Water, Sources, Health Risk, and Remediation Strategies: An Overview. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E5438. [PMID: 32731582 PMCID: PMC7432837 DOI: 10.3390/ijerph17155438] [Citation(s) in RCA: 166] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 01/23/2023]
Abstract
Chromium is a potentially toxic metal occurring in water and groundwater as a result of natural and anthropogenic sources. Microbial interaction with mafic and ultramafic rocks together with geogenic processes release Cr (VI) in natural environment by chromite oxidation. Moreover, Cr (VI) pollution is largely related to several Cr (VI) industrial applications in the field of energy production, manufacturing of metals and chemicals, and subsequent waste and wastewater management. Chromium discharge in European Union (EU) waters is subjected to nationwide recommendations, which vary depending on the type of industry and receiving water body. Once in water, chromium mainly occurs in two oxidation states Cr (III) and Cr (VI) and related ion forms depending on pH values, redox potential, and presence of natural reducing agents. Public concerns with chromium are primarily related to hexavalent compounds owing to their toxic effects on humans, animals, plants, and microorganisms. Risks for human health range from skin irritation to DNA damages and cancer development, depending on dose, exposure level, and duration. Remediation strategies commonly used for Cr (VI) removal include physico-chemical and biological methods. This work critically presents their advantages and disadvantages, suggesting a site-specific and accurate evaluation for choosing the best available recovering technology.
Collapse
Affiliation(s)
- Marina Tumolo
- Water Research, Institute-Italian National Research Council (IRSA-CNR), 70132 Bari, Italy; (M.T.); (D.L.); (C.C.); (C.M.); (V.F.U.)
- Department of Biology, University of Bari, 70126 Bari, Italy
| | - Valeria Ancona
- Water Research, Institute-Italian National Research Council (IRSA-CNR), 70132 Bari, Italy; (M.T.); (D.L.); (C.C.); (C.M.); (V.F.U.)
| | - Domenico De Paola
- Institute of Biosciences and Bioresources, Italian National Research Council (IBBR-CNR), 70126 Bari, Italy;
| | - Daniela Losacco
- Water Research, Institute-Italian National Research Council (IRSA-CNR), 70132 Bari, Italy; (M.T.); (D.L.); (C.C.); (C.M.); (V.F.U.)
- Department of Biology, University of Bari, 70126 Bari, Italy
| | - Claudia Campanale
- Water Research, Institute-Italian National Research Council (IRSA-CNR), 70132 Bari, Italy; (M.T.); (D.L.); (C.C.); (C.M.); (V.F.U.)
| | - Carmine Massarelli
- Water Research, Institute-Italian National Research Council (IRSA-CNR), 70132 Bari, Italy; (M.T.); (D.L.); (C.C.); (C.M.); (V.F.U.)
| | - Vito Felice Uricchio
- Water Research, Institute-Italian National Research Council (IRSA-CNR), 70132 Bari, Italy; (M.T.); (D.L.); (C.C.); (C.M.); (V.F.U.)
| |
Collapse
|
23
|
Luczak MW, Krawic C, Zhitkovich A. p53 activation by Cr(VI): a transcriptionally limited response induced by ATR kinase in S-phase. Toxicol Sci 2019; 172:11-22. [PMID: 31388677 PMCID: PMC6813752 DOI: 10.1093/toxsci/kfz178] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 07/19/2019] [Accepted: 07/26/2019] [Indexed: 01/30/2023] Open
Abstract
Cellular reduction of carcinogenic chromium(VI) causes several forms of Cr-DNA damage with different genotoxic properties. Chromate-treated cultured cells have shown a strong proapoptotic activity of the DNA damage-sensitive transcription factor p53. However, induction of p53 transcriptional targets by Cr(VI) in rodent lungs was weak or undetectable. We examined Cr(VI) effects on the p53 pathway in human cells with restored levels of ascorbate that acts as a principal reducer of Cr(VI) in vivo but is nearly absent in standard cell cultures. Ascorbate-restored H460 and primary human cells treated with Cr(VI) contained higher levels of p53 and its Ser15 phosphorylation, which were induced by ATR kinase. Cr(VI)-stimulated p53 phosphorylation occurred in S-phase by a diffusible pool of ATR that was separate from the chromatin-bound pool targeting DNA repair substrates at the sites of toxic mismatch repair of Cr-DNA adducts. Even when more abundantly present than after exposure to the radiomimetic bleomycin, Cr(VI)-stabilized p53 showed a much more limited activation of its target genes in two types of primary human cells. No increases in mRNA were found for nucleotide excision repair factors and a majority of proapoptotic genes. A weak transcription activity of Cr(VI)-upregulated p53 was associated with its low lysine acetylation in the regulatory C-terminal domain, resulting from the inability of Cr(VI) to activate ATM in ascorbate-restored cells. Thus, p53 activation by ascorbate-metabolized Cr(VI) represents a limited genome-protective response that is defective in upregulation of DNA repair genes and proapoptotic transcripts for elimination of damaged cells.
Collapse
Affiliation(s)
- Michal W Luczak
- Brown University, Department of Pathology and Laboratory Medicine, Providence, RI, USA
| | - Casey Krawic
- Brown University, Department of Pathology and Laboratory Medicine, Providence, RI, USA
| | - Anatoly Zhitkovich
- Brown University, Department of Pathology and Laboratory Medicine, Providence, RI, USA
| |
Collapse
|
24
|
Chen QY, Murphy A, Sun H, Costa M. Molecular and epigenetic mechanisms of Cr(VI)-induced carcinogenesis. Toxicol Appl Pharmacol 2019; 377:114636. [PMID: 31228494 DOI: 10.1016/j.taap.2019.114636] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/17/2019] [Accepted: 06/18/2019] [Indexed: 12/11/2022]
Abstract
Chromium (Cr) is a naturally occurring metallic element found in the Earth's crust. While trivalent chromium ([Cr(III)] is considered non-carcinogenic, hexavalent chromium [Cr(VI)] has long been established as an IARC class I human carcinogen, known to induce cancers of the lung. Current literature suggests that Cr(VI) is capable of inducing carcinogenesis through both genetic and epigenetic mechanisms. Although much has been learned about the molecular etiology of Cr(VI)-induced lung carcinogenesis, more remains to be explored. In particular, the explicit epigenetic alterations induced by Cr(VI) in lung cancer including histone modifications and miRNAs, remain understudied. Through comprehensive review of available literature found between 1973 and 2019, this article provides a summary of updated understanding of the molecular mechanisms of Cr(VI)-carcinogenesis. In addition, this review identifies potential research gaps in the areas of histone modifications and miRNAs, which may prompt new niches for future research.
Collapse
Affiliation(s)
- Qiao Yi Chen
- Department of Environmental Medicine, New York University School of Medicine, 341 East 25 Street, New York, NY 10016, United States of America.
| | - Anthony Murphy
- Department of Environmental Medicine, New York University School of Medicine, 341 East 25 Street, New York, NY 10016, United States of America.
| | - Hong Sun
- Department of Environmental Medicine, New York University School of Medicine, 341 East 25 Street, New York, NY 10016, United States of America.
| | - Max Costa
- Department of Environmental Medicine, New York University School of Medicine, 341 East 25 Street, New York, NY 10016, United States of America.
| |
Collapse
|
25
|
Zablon HA, VonHandorf A, Puga A. Chromium exposure disrupts chromatin architecture upsetting the mechanisms that regulate transcription. Exp Biol Med (Maywood) 2019; 244:752-757. [PMID: 30935235 PMCID: PMC6567585 DOI: 10.1177/1535370219839953] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
IMPACT STATEMENT This mini-review highlights current evidence on the mechanisms through which hexavalent chromium (Cr(VI)) disrupts transcriptional regulation, an emerging area of interest and one of the central processes by which chromium induces carcinogenesis. Several studies have shown that Cr(VI) causes widespread DNA damage and disrupts epigenetic signatures, suggesting that chromatin may be a direct Cr(VI) target. The findings discussed here suggest that Cr(VI) disrupts transcriptional regulation by causing genomic architecture changes.
Collapse
Affiliation(s)
- Hesbon A Zablon
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Andrew VonHandorf
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Alvaro Puga
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
26
|
Rubis B, Luczak MW, Krawic C, Zhitkovich A. Vitamin C increases DNA breaks and suppresses DNA damage-independent activation of ATM by bleomycin. Free Radic Biol Med 2019; 136:12-21. [PMID: 30926564 PMCID: PMC6488359 DOI: 10.1016/j.freeradbiomed.2019.03.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/25/2019] [Accepted: 03/25/2019] [Indexed: 01/07/2023]
Abstract
Bleomycin is a redox-active drug with anticancer and other clinical applications. It is also frequently used as a tool in fundamental research on cellular responses to DNA double-strand breaks (DSBs). A conversion of bleomycin into its DNA-breaking form requires Fe, one-electron donors and O2. Here, we examined how a major biological antioxidant ascorbate (reduced vitamin C), which is practically absent in standard cell culture, impacts cellular responses to bleomycin. We found that restoration of physiological levels of vitamin C in human cancer cells increased their killing by bleomycin in 2D cultures and 3D tumor spheroids. Higher cytotoxicity of bleomycin occurred in cells with normal and shRNA-depleted p53. Cellular vitamin C enhanced the ability of bleomycin by produce DSBs, which was established by direct measurements of these lesions in three cell lines. Vitamin C-restored cancer cells also showed a higher sensitivity to killing by low-dose bleomycin in combination with inhibitors of DSB repair-activating ATM or DNA-PK kinases. The presence of ascorbate in bleomycin-treated cells suppressed a DSB-independent activation of the ATM-CHK2 axis by blocking superoxide radical. In vitro studies detected a greatly superior ability of ascorbate over other cellular reducers to catalyze DSB formation by bleomycin. Ascorbate was faster than other antioxidants in promoting two steps in activation of bleomycin. Our results demonstrate strong activation effects of vitamin C on bleomycin, shifting its toxicity further toward DNA damage and making it more sensitive to manipulations of DNA repair.
Collapse
Affiliation(s)
- Blazej Rubis
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship Street, Providence, RI, 02912, USA
| | - Michal W Luczak
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship Street, Providence, RI, 02912, USA
| | - Casey Krawic
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship Street, Providence, RI, 02912, USA
| | - Anatoly Zhitkovich
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship Street, Providence, RI, 02912, USA.
| |
Collapse
|
27
|
Roles of Bacillus subtilis RecA, Nucleotide Excision Repair, and Translesion Synthesis Polymerases in Counteracting Cr(VI)-Promoted DNA Damage. J Bacteriol 2019; 201:JB.00073-19. [PMID: 30745368 DOI: 10.1128/jb.00073-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 01/29/2019] [Indexed: 11/20/2022] Open
Abstract
Bacteria deploy global programs of gene expression, including components of the SOS response, to counteract the cytotoxic and genotoxic effects of environmental DNA-damaging factors. Here we report that genetic damage promoted by hexavalent chromium elicited the SOS response in Bacillus subtilis, as evidenced by the induction of transcriptional uvrA-lacZ, recA-lacZ, and P recA-gfp fusions. Accordingly, B. subtilis strains deficient in homologous recombination (RecA) and nucleotide excision repair (NER) (UvrA), components of the SOS response, were significantly more sensitive to Cr(VI) treatment than were cells of the wild-type strain. These results strongly suggest that Cr(VI) induces the formation in growing B. subtilis cells of cytotoxic and genotoxic bulky DNA lesions that are processed by RecA and/or the NER pathways. In agreement with this notion, Cr(VI) significantly increased the formation of DNA-protein cross-links (DPCs) and induced mutagenesis in recA- and uvrA-deficient B. subtilis strains, through a pathway that required YqjH/YqjW-mediated translesion synthesis. We conclude that Cr(VI) promotes mutagenesis and cell death in B. subtilis by a mechanism that involves the formation of DPCs and that such deleterious effects are counteracted by both the NER and homologous recombination pathways, belonging to the RecA-dependent SOS system.IMPORTANCE It has been shown that, following permeation of cell barriers, Cr(VI) kills B. subtilis cells following a mechanism of reactive oxygen species-promoted DNA damage, which is counteracted by the guanine oxidized repair system. Here we report a distinct mechanism of Cr(VI)-promoted DNA damage that involves production of DPCs capable of eliciting the bacterial SOS response. We also report that the NER and homologous recombination (RecA) repair pathways, as well as low-fidelity DNA polymerases, counteract this metal-induced mechanism of killing in B. subtilis Hence, our results contribute to an understanding of how environmental pollutants activate global programs of gene expression that allow bacteria to contend with the cytotoxic and genotoxic effects of heavy metals.
Collapse
|
28
|
Krawic C, Zhitkovich A. Toxicological Antagonism among Welding Fume Metals: Inactivation of Soluble Cr(VI) by Iron. Chem Res Toxicol 2018; 31:1172-1184. [PMID: 30362728 PMCID: PMC6247247 DOI: 10.1021/acs.chemrestox.8b00182] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Indexed: 12/19/2022]
Abstract
Epidemiological studies in chromate production have established hexavalent chromium as a potent lung carcinogen. Inhalation of chromium(VI) most often occurs in mixtures with other metals as among stainless steel welders, which is the largest occupational group with Cr(VI) exposure. Surprisingly, carcinogenicity of Cr(VI)-containing welding fumes is moderate and not consistently higher than that of Cr-free welding. Here, we investigated interactions between chromate and three other metal ions [Fe(III), Mn(II), Ni(II)] that are typically released from stainless steel welding particles. In human lung epithelial cells with physiological levels of ascorbate and glutathione, Cr(VI) was by far the most cytotoxic metal in single exposures. Coexposure with Fe(III) suppressed cytotoxicity and genotoxicity of Cr(VI), which resulted from a severe inhibition of Cr uptake by cells and required extracellular ascorbate/glutathione. Chemically, detoxification of Cr(VI) occurred via its rapid extracellular reduction by Fe(II) that primarily originated from ascorbate-reduced Fe(III). Glutathione was a significant contributor to reduction of Cr(VI) by Fe only in the presence of ascorbate. We further found that variability in Cr(VI) metabolism among common cell culture media was caused by their different Fe content. Ni(II) and Mn(II) had no detectable effects on metabolism, cellular uptake or cytotoxicity of Cr(VI). The main biological findings were confirmed in three human lung cell lines, including stem cell-like and primary cells. We discovered extracellular detoxification of carcinogenic chromate in coexposures with Fe(III) ions and identified the underlying chemical mechanism. Our findings established an important case when exposure to mixtures causes inactivation of a potent human carcinogen.
Collapse
Affiliation(s)
- Casey Krawic
- Department of Pathology and Laboratory
Medicine, Brown University, 70 Ship Street, Providence, Rhode Island 02912, United States
| | - Anatoly Zhitkovich
- Department of Pathology and Laboratory
Medicine, Brown University, 70 Ship Street, Providence, Rhode Island 02912, United States
| |
Collapse
|
29
|
Luczak MW, Zhitkovich A. Monoubiquitinated γ-H2AX: Abundant product and specific biomarker for non-apoptotic DNA double-strand breaks. Toxicol Appl Pharmacol 2018; 355:238-246. [PMID: 30006243 PMCID: PMC6754567 DOI: 10.1016/j.taap.2018.07.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 06/23/2018] [Accepted: 07/09/2018] [Indexed: 02/07/2023]
Abstract
DNA double-strand breaks (DSBs) are a highly toxic form of DNA damage produced by a number of carcinogens, drugs, and metabolic abnormalities. Involvement of DSBs in many pathologies has led to frequent measurements of these lesions, primarily via biodosimetry of S139-phosphorylated histone H2AX (γ-H2AX). However, γ-H2AX is also induced by some non-DSB conditions and abundantly formed in apoptosis, raising concerns about the overestimation of potential genotoxic agents and accuracy of DSB assessments. DSB-triggered γ-H2AX undergoes RNF168-mediated K13/K15 monoubiquitination, which is rarely analyzed in DSB/genotoxicity studies. Here we identified critical methodological factors that are necessary for the efficient detection of mono- (ub1) and diubiquitinated (ub2) γ-H2AX. Using optimized technical conditions, we found that γ-H2AX-ub1 was a predominant form of γ-H2AX in three primary human cell lines containing mechanistically distinct types of DSBs. Replication stress-associated DSBs also triggered extensive formation of γ-H2AX-ub1. For DSBs induced by oxidative damage or topoisomerase II, both γ-H2AX and γ-H2AX-ub1 showed dose-dependent increases whereas γ-H2AX-ub2 plateaued at low levels of breaks. Despite abundance of γ-H2AX, γ-H2AX-ub1,2 formation was blocked in apoptosis, which was associated with proteolytic cleavage of RNF168. Chromatin damage also caused only the production of γ-H2AX but not its ub1,2 forms. Our results revealed a major contribution of ubiquitinated forms to the overall γ-H2AX response and demonstrated the specificity of monoubiquitinated γ-H2AX as a biodosimeter of non-apoptotic DSBs.
Collapse
Affiliation(s)
- Michal W Luczak
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02912, USA
| | - Anatoly Zhitkovich
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
30
|
Tdp1 processes chromate-induced single-strand DNA breaks that collapse replication forks. PLoS Genet 2018; 14:e1007595. [PMID: 30148840 PMCID: PMC6128646 DOI: 10.1371/journal.pgen.1007595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 09/07/2018] [Accepted: 07/26/2018] [Indexed: 01/20/2023] Open
Abstract
Hexavalent chromium [Cr(VI)] damages DNA and causes cancer, but it is unclear which DNA damage responses (DDRs) most critically protect cells from chromate toxicity. Here, genome-wide quantitative functional profiling, DDR measurements and genetic interaction assays in Schizosaccharomyces pombe reveal a chromate toxicogenomic profile that closely resembles the cancer chemotherapeutic drug camptothecin (CPT), which traps Topoisomerase 1 (Top1)-DNA covalent complex (Top1cc) at the 3’ end of single-stand breaks (SSBs), resulting in replication fork collapse. ATR/Rad3-dependent checkpoints that detect stalled and collapsed replication forks are crucial in Cr(VI)-treated cells, as is Mus81-dependent sister chromatid recombination (SCR) that repairs single-ended double-strand breaks (seDSBs) at broken replication forks. Surprisingly, chromate resistance does not require base excision repair (BER) or interstrand crosslink (ICL) repair, nor does co-elimination of XPA-dependent nucleotide excision repair (NER) and Rad18-mediated post-replication repair (PRR) confer chromate sensitivity in fission yeast. However, co-elimination of Tdp1 tyrosyl-DNA phosphodiesterase and Rad16-Swi10 (XPF-ERCC1) NER endonuclease synergistically enhances chromate toxicity in top1Δ cells. Pnk1 polynucleotide kinase phosphatase (PNKP), which restores 3’-hydroxyl ends to SSBs processed by Tdp1, is also critical for chromate resistance. Loss of Tdp1 ameliorates pnk1Δ chromate sensitivity while enhancing the requirement for Mus81. Thus, Tdp1 and PNKP, which prevent neurodegeneration in humans, repair an important class of Cr-induced SSBs that collapse replication forks. Hexavalent chromium is a carcinogen that is found at toxic waste sites and in some groundwater supplies. Cellular metabolism converts chromium into DNA-damaging chromate, but it is unclear which types of chromate-DNA lesions are most dangerous, and which cellular mechanisms most critically prevent chromium toxicity. This study uses whole-genome profiling to identify DNA repair pathways that are crucial for chromate resistance in fission yeast. The resulting ‘toxicogenomic’ profile of chromate closely matches camptothecin, a natural product representing a class of chemotherapeutic drugs that cause replication fork collapse by poisoning Topoisomerase 1 (Top1), which relaxes supercoiled DNA by creating and resealing single-strand breaks (SSBs). Genetic interaction analyses uncover important roles for Tdp1 tyrosyl-DNA phosphodiesterase and Pnk1 polynucleotide 5’-kinase 3’-phosphatase (PNKP), which repair camptothecin-induced SSBs and prevent neurological disease in humans. However, chromium toxicity does not involve Top1. As Tdp1 and Pnk1 repair SSBs with 3’-blocked termini, these data suggest that Top1-independent 3’-blocked SSBs contribute to the carcinogenic and mutagenic properties of chromium.
Collapse
|
31
|
Wang L, Xu Z, Khawar MB, Liu C, Li W. The histone codes for meiosis. Reproduction 2018; 154:R65-R79. [PMID: 28696245 DOI: 10.1530/rep-17-0153] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 06/10/2017] [Accepted: 06/19/2017] [Indexed: 12/28/2022]
Abstract
Meiosis is a specialized process that produces haploid gametes from diploid cells by a single round of DNA replication followed by two successive cell divisions. It contains many special events, such as programmed DNA double-strand break (DSB) formation, homologous recombination, crossover formation and resolution. These events are associated with dynamically regulated chromosomal structures, the dynamic transcriptional regulation and chromatin remodeling are mainly modulated by histone modifications, termed 'histone codes'. The purpose of this review is to summarize the histone codes that are required for meiosis during spermatogenesis and oogenesis, involving meiosis resumption, meiotic asymmetric division and other cellular processes. We not only systematically review the functional roles of histone codes in meiosis but also discuss future trends and perspectives in this field.
Collapse
Affiliation(s)
- Lina Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Zhiliang Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China.,Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | | | - Chao Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
| |
Collapse
|
32
|
Pustovalova M, Astrelina ТA, Grekhova A, Vorobyeva N, Tsvetkova A, Blokhina T, Nikitina V, Suchkova Y, Usupzhanova D, Brunchukov V, Kobzeva I, Karaseva Т, Ozerov IV, Samoylov A, Bushmanov A, Leonov S, Izumchenko E, Zhavoronkov A, Klokov D, Osipov AN. Residual γH2AX foci induced by low dose x-ray radiation in bone marrow mesenchymal stem cells do not cause accelerated senescence in the progeny of irradiated cells. Aging (Albany NY) 2018; 9:2397-2410. [PMID: 29165316 PMCID: PMC5723693 DOI: 10.18632/aging.101327] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/11/2017] [Indexed: 01/09/2023]
Abstract
Mechanisms underlying the effects of low-dose ionizing radiation (IR) exposure (10-100 mGy) remain unknown. Here we present a comparative study of early (less than 24h) and delayed (up to 11 post-irradiation passages) radiation effects caused by low (80 mGy) vs intermediate (1000 mGy) dose X-ray exposure in cultured human bone marrow mesenchymal stem cells (MSCs). We show that γН2АХ foci induced by an intermediate dose returned back to the control value by 24 h post-irradiation. In contrast, low-dose irradiation resulted in residual γН2АХ foci still present at 24 h. Notably, these low dose induced residual γН2АХ foci were not co-localized with рАТМ foci and were observed predominantly in the proliferating Кi67 positive (Кi67+) cells. The number of γН2АХ foci and the fraction of nonproliferating (Кi67-) and senescent (SA-β-gal+) cells measured at passage 11 were increased in cultures exposed to an intermediate dose compared to unirradiated controls. These delayed effects were not seen in the progeny of cells that were irradiated with low-dose X-rays, although such exposure resulted in residual γН2АХ foci in directly irradiated cells. Taken together, our results support the hypothesis that the low-dose IR induced residual γH2AХ foci do not play a role in delayed irradiation consequences, associated with cellular senescence in cultured MSCs.
Collapse
Affiliation(s)
- Margarita Pustovalova
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia.,Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Тatiana A Astrelina
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia
| | - Anna Grekhova
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia.,Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow 119991, Russia.,Emanuel Institute for Biochemical Physics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Natalia Vorobyeva
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia.,Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Anastasia Tsvetkova
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia.,Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region 141700, Russia
| | - Taisia Blokhina
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia.,Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Victoria Nikitina
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia
| | - Yulia Suchkova
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia
| | - Daria Usupzhanova
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia
| | - Vitalyi Brunchukov
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia
| | - Irina Kobzeva
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia
| | - Тatiana Karaseva
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia
| | - Ivan V Ozerov
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia.,Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Aleksandr Samoylov
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia
| | - Andrey Bushmanov
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia
| | - Sergey Leonov
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region 141700, Russia.,Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Evgeny Izumchenko
- Department of Otolaryngology-Head and Neck Cancer Research, Johns Hopkins University, School of Medicine, Baltimore, MD 21218, USA
| | - Alex Zhavoronkov
- Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Dmitry Klokov
- Canadian Nuclear Laboratories, Chalk River, Ontario K0J1P0, Canada.,University of Ottawa, Ottawa, Ontario K1N6N5, Canada
| | - Andreyan N Osipov
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia.,Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow 119991, Russia.,Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region 141700, Russia.,Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
33
|
Levina A, Crans DC, Lay PA. Speciation of metal drugs, supplements and toxins in media and bodily fluids controls in vitro activities. Coord Chem Rev 2017. [DOI: 10.1016/j.ccr.2017.01.002] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|
34
|
Thompson CM, Suh M, Proctor DM, Haws LC, Harris MA. Ten factors for considering the mode of action of Cr(VI)-induced gastrointestinal tumors in rodents. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2017; 823:45-57. [DOI: 10.1016/j.mrgentox.2017.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 08/26/2017] [Accepted: 08/28/2017] [Indexed: 12/28/2022]
|
35
|
Krawic C, Luczak MW, Zhitkovich A. Variation in Extracellular Detoxification Is a Link to Different Carcinogenicity among Chromates in Rodent and Human Lungs. Chem Res Toxicol 2017; 30:1720-1729. [PMID: 28759204 PMCID: PMC5605882 DOI: 10.1021/acs.chemrestox.7b00172] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Indexed: 11/30/2022]
Abstract
Inhalation of soluble chromium(VI) is firmly linked with higher risks of lung cancer in humans. However, comparative studies in rats have found a high lung tumorigenicity for moderately soluble chromates but no tumors for highly soluble chromates. These major species differences remain unexplained. We investigated the impact of extracellular reducers on responses of human and rat lung epithelial cells to different Cr(VI) forms. Extracellular reduction of Cr(VI) is a detoxification process, and rat and human lung lining fluids contain different concentrations of ascorbate and glutathione. We found that reduction of chromate anions in simulated lung fluids was principally driven by ascorbate with only minimal contribution from glutathione. The addition of 500 μM ascorbate (∼rat lung fluid concentration) to culture media strongly inhibited cellular uptake of chromate anions and completely prevented their cytotoxicity even at otherwise lethal doses. While proportionally less effective, 50 μM extracellular ascorbate (∼human lung fluid concentration) also decreased uptake of chromate anions and their cytotoxicity. In comparison to chromate anions, uptake and cytotoxicity of respirable particles of moderately soluble CaCrO4 and SrCrO4 were much less sensitive to suppression by extracellular ascorbate, especially during early exposure times and in primary bronchial cells. In the absence of extracellular ascorbate, chromate anions and CaCrO4/SrCrO4 particles produced overall similar levels of DNA double-stranded breaks, with less soluble particles exhibiting a slower rate of breakage. Our results indicate that a gradual extracellular dissolution and a rapid internalization of calcium chromate and strontium chromate particles makes them resistant to detoxification outside the cells, which is extremely effective for chromate anions in the rat lung fluid. The detoxification potential of the human lung fluid is significant but much lower and insufficient to provide a threshold-type dose dependence for soluble chromates.
Collapse
Affiliation(s)
- Casey Krawic
- Department of Pathology and
Laboratory Medicine, Brown University, 70 Ship Street, Providence, Rhode Island 02912, United States
| | - Michal W. Luczak
- Department of Pathology and
Laboratory Medicine, Brown University, 70 Ship Street, Providence, Rhode Island 02912, United States
| | - Anatoly Zhitkovich
- Department of Pathology and
Laboratory Medicine, Brown University, 70 Ship Street, Providence, Rhode Island 02912, United States
| |
Collapse
|
36
|
Luczak MW, Zhitkovich A. Nickel-induced HIF-1α promotes growth arrest and senescence in normal human cells but lacks toxic effects in transformed cells. Toxicol Appl Pharmacol 2017; 331:94-100. [PMID: 28552779 DOI: 10.1016/j.taap.2017.05.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 05/18/2017] [Accepted: 05/24/2017] [Indexed: 02/09/2023]
Abstract
Nickel is a human carcinogen that acts as a hypoxia mimic by activating the transcription factor HIF-1α and hypoxia-like transcriptomic responses. Hypoxia and elevated HIF-1α are typically associated with drug resistance in cancer cells, which is caused by increased drug efflux and other mechanisms. Here we examined the role of HIF-1α in uptake of soluble Ni(II) and Ni(II)-induced cell fate outcomes using si/shRNA knockdowns and gene deletion models. We found that HIF-1α had no effect on accumulation of Ni(II) in two transformed (H460, A549) and two normal human cell lines (IMR90, WI38). The loss of HIF-1α also produced no significant impact on p53-dependent and p53-independent apoptotic responses or clonogenic survival of Ni(II)-treated transformed cells. In normal human cells, HIF-1α enhanced the ability of Ni(II) to inhibit cell proliferation and cause a permanent growth arrest (senescence). Consistent with its growth-suppressive effects, HIF-1α was important for upregulation of the cell cycle inhibitors p21 (CDKN1A) and p27 (CDKN1B). Irrespective of HIF-1α status, Ni(II) strongly increased levels of MYC protein but did not change protein expression of the cell cycle-promoting phosphatase CDC25A or the CDK inhibitor p16. Our findings indicate that HIF-1α limits propagation of Ni(II)-damaged normal cells, suggesting that it may act in a tumor suppressor-like manner during early stages of Ni(II) carcinogenesis.
Collapse
Affiliation(s)
- Michal W Luczak
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02912, USA
| | - Anatoly Zhitkovich
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
37
|
Ortega-Atienza S, Krawic C, Watts L, McCarthy C, Luczak MW, Zhitkovich A. 20S immunoproteasomes remove formaldehyde-damaged cytoplasmic proteins suppressing caspase-independent cell death. Sci Rep 2017; 7:654. [PMID: 28381880 PMCID: PMC5429636 DOI: 10.1038/s41598-017-00757-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/09/2017] [Indexed: 01/08/2023] Open
Abstract
Immunoproteasomes are known for their involvement in antigen presentation. However, their broad tissue presence and other evidence are indicative of nonimmune functions. We examined a role for immunoproteasomes in cellular responses to the endogenous and environmental carcinogen formaldehyde (FA) that binds to cytosolic and nuclear proteins producing proteotoxic stress and genotoxic DNA-histone crosslinks. We found that immunoproteasomes were important for suppression of a caspase-independent cell death and the long-term survival of FA-treated cells. All major genotoxic responses to FA, including replication inhibition and activation of the transcription factor p53 and the apical ATM and ATR kinases, were unaffected by immunoproteasome inactivity. Immunoproteasome inhibition enhanced activation of the cytosolic protein damage sensor HSF1, elevated levels of K48-polyubiquitinated cytoplasmic proteins and increased depletion of unconjugated ubiquitin. We further found that FA induced the disassembly of 26S immunoproteasomes, but not standard 26S proteasomes, releasing the 20S catalytic immunoproteasome. FA-treated cells also had higher amounts of small activators PA28αβ and PA28γ bound to 20S particles. Our findings highlight the significance of nonnuclear damage in FA injury and reveal a major role for immunoproteasomes in elimination of FA-damaged cytoplasmic proteins through ubiquitin-independent proteolysis.
Collapse
Affiliation(s)
- Sara Ortega-Atienza
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Casey Krawic
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Lauren Watts
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Caitlin McCarthy
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Michal W Luczak
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Anatoly Zhitkovich
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA.
| |
Collapse
|
38
|
Vimercati L, Gatti MF, Gagliardi T, Cuccaro F, De Maria L, Caputi A, Quarato M, Baldassarre A. Environmental exposure to arsenic and chromium in an industrial area. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2017; 24:11528-11535. [PMID: 28321698 PMCID: PMC5393286 DOI: 10.1007/s11356-017-8827-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 03/13/2017] [Indexed: 05/06/2023]
Abstract
Arsenic and chromium are widespread environmental contaminants that affect global health due to their toxicity and carcinogenicity. To date, few studies have investigated exposure to arsenic and chromium in a population residing in a high-risk environmental area. The aim of this study is to evaluate the exposure to arsenic and chromium in the general population with no occupational exposure to these metals, resident in the industrial area of Taranto, Southern Italy, through biological monitoring techniques. We measured the levels of chromium, inorganic arsenic, and methylated metabolites, in the urine samples of 279 subjects residing in Taranto and neighboring areas. Qualified health staff administered a standardized structured questionnaire investigating lifestyle habits and controlling for confounding factors. The biological monitoring data showed high urinary concentrations of both the heavy metals investigated, particularly Cr. On this basis, it will be necessary to carry out an organized environmental monitoring program, taking into consideration all exposure routes so as to correlate the environmental concentrations of these metals with the biomonitoring results.
Collapse
Affiliation(s)
- Luigi Vimercati
- Interdisciplinary Department of Medicine, Occupational Medicine “B. Ramazzini”, University of Bari Medical School, Giulio Cesare Square 11, 70124 Bari, Italy
| | - Maria F Gatti
- Interdisciplinary Department of Medicine, Occupational Medicine “B. Ramazzini”, University of Bari Medical School, Giulio Cesare Square 11, 70124 Bari, Italy
| | - Tommaso Gagliardi
- Interdisciplinary Department of Medicine, Occupational Medicine “B. Ramazzini”, University of Bari Medical School, Giulio Cesare Square 11, 70124 Bari, Italy
| | | | - Luigi De Maria
- Interdisciplinary Department of Medicine, Occupational Medicine “B. Ramazzini”, University of Bari Medical School, Giulio Cesare Square 11, 70124 Bari, Italy
| | - Antonio Caputi
- Interdisciplinary Department of Medicine, Occupational Medicine “B. Ramazzini”, University of Bari Medical School, Giulio Cesare Square 11, 70124 Bari, Italy
| | - Marco Quarato
- Interdisciplinary Department of Medicine, Occupational Medicine “B. Ramazzini”, University of Bari Medical School, Giulio Cesare Square 11, 70124 Bari, Italy
| | - Antonio Baldassarre
- Interdisciplinary Department of Medicine, Occupational Medicine “B. Ramazzini”, University of Bari Medical School, Giulio Cesare Square 11, 70124 Bari, Italy
| |
Collapse
|
39
|
Gene 33/Mig6 inhibits hexavalent chromium-induced DNA damage and cell transformation in human lung epithelial cells. Oncotarget 2017; 7:8916-30. [PMID: 26760771 PMCID: PMC4891014 DOI: 10.18632/oncotarget.6866] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/25/2015] [Indexed: 01/01/2023] Open
Abstract
Hexavalent Chromium [Cr(VI)] compounds are human lung carcinogens and environmental/occupational hazards. The molecular mechanisms of Cr(VI) carcinogenesis appear to be complex and are poorly defined. In this study, we investigated the potential role of Gene 33 (ERRFI1, Mig6), a multifunctional adaptor protein, in Cr(VI)-mediated lung carcinogenesis. We show that the level of Gene 33 protein is suppressed by both acute and chronic Cr(VI) treatments in a dose- and time-dependent fashion in BEAS-2B lung epithelial cells. The inhibition also occurs in A549 lung bronchial carcinoma cells. Cr(VI) suppresses Gene 33 expression mainly through post-transcriptional mechanisms, although the mRNA level of gene 33 also tends to be lower upon Cr(VI) treatments. Cr(VI)-induced DNA damage appears primarily in the S phases of the cell cycle despite the high basal DNA damage signals at the G2M phase. Knockdown of Gene 33 with siRNA significantly elevates Cr(VI)-induced DNA damage in both BEAS-2B and A549 cells. Depletion of Gene 33 also promotes Cr(VI)-induced micronucleus (MN) formation and cell transformation in BEAS-2B cells. Our results reveal a novel function of Gene 33 in Cr(VI)-induced DNA damage and lung epithelial cell transformation. We propose that in addition to its role in the canonical EGFR signaling pathway and other signaling pathways, Gene 33 may also inhibit Cr(VI)-induced lung carcinogenesis by reducing DNA damage triggered by Cr(VI).
Collapse
|
40
|
Bruno M, Ross J, Ge Y. Proteomic responses of BEAS-2B cells to nontoxic and toxic chromium: Protein indicators of cytotoxicity conversion. Toxicol Lett 2016; 264:59-70. [PMID: 27592090 DOI: 10.1016/j.toxlet.2016.08.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 08/25/2016] [Accepted: 08/30/2016] [Indexed: 12/25/2022]
Abstract
Hexavalent chromium (Cr (VI)) is an environmental human carcinogen which primarily targets lungs. Among a variety of toxic mechanisms, disruption of biological pathways via translational and post-translational modifications represents a key mechanism through which Cr (VI) induces cytotoxicity and carcinogenesis. To identify those disruptions which are altered in response to cytotoxic Cr (VI) exposures, we measured and compared cytotoxicity and changes in expression and phosphorylation status of 15 critical biochemical pathway regulators in human BEAS-2B cells exposed for 48h to a non-toxic concentration (0.3μM) and a toxic concentration (1.8μM) of Cr (VI) by ELISA techniques. In addition, 43 functional proteins which may be altered in response to pathway signaling changes were identified using two dimensional electrophoresis (2-DE) and mass spectrometry. The proteins and fold changes observed in cells exposed to the non-toxic dose of Cr (VI) (0.3μM) were not necessarily the same as those found in the toxic one (1.8μM). A subset of signaling proteins that were correlated with the cytotoxic responses of human BEAS-2B cells to Cr (VI) treatments were identified. These proteins include regulators of glycolysis, glycogen synthase kinase 3 beta (GSK3β) and phosphoprotein 70 ribosomal protein s6 kinase (p70S6K), a signaling protein associated with oxidative stress and inflammation responses, JNK and metal regulatory transcription factor 1 (MTF-1), and a source of ubiquitin for signaling targeted protein degradation, polyubiquitin C (UBC). In addition, two dimensional gel electrophoresis (2-DE) was applied to identify key alterations in biochemical pathways differentiating between cytotoxic and non-cytotoxic exposures to Cr (VI), including glycolysis and gluconeogenesis, protein degradation, inflammation, and oxidative stress.
Collapse
Affiliation(s)
- Maribel Bruno
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Jeffrey Ross
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Yue Ge
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| |
Collapse
|
41
|
Van Doorslaer S, Beirinckx Q, Nys K, Mangiameli MF, Cuypers B, Callens F, Vrielinck H, González JC. EPR and DFT analysis of biologically relevant chromium(V) complexes with d-glucitol and d-glucose. J Inorg Biochem 2016; 162:216-226. [PMID: 27460210 DOI: 10.1016/j.jinorgbio.2016.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 07/15/2016] [Accepted: 07/18/2016] [Indexed: 10/21/2022]
Abstract
1,2-diolato ligands, such as carbohydrates and glycoproteins, tend to stabilize chromium(V), thus forming important intermediates that have been implicated in the genotoxicity of Cr(VI). Since many years, room-temperature continuous-wave electron paramagnetic resonance (EPR) at X-band microwave frequencies has been used as a standard characterization tool to study chromium(V) intermediates formed during the reduction of Cr(VI) in the presence of biomolecules. In this work, the added value is tested of using a combination of pulsed and high-field EPR techniques with density functional theory computations to unravel the nature of Cr(V) complexes with biologically relevant chelators, such as carbohydrates. The study focuses on the oxidochromium(V) complexes formed during reduction of potassium dichromate with glutathione in the presence of the monosaccharide d-glucose or the polyalcohol d-glucitol. It is shown that although the presence of a multitude of Cr(V) intermediates may hamper a complete structural determination, the combined EPR and DFT approach reveals unambiguously the effect of freezing on the location of the counterions, the gradual replacement of water ligands by the diols, and the preference of Cr(V) to bind certain conformers.
Collapse
Affiliation(s)
- Sabine Van Doorslaer
- University of Antwerp, Department of Physics, Universiteitsplein 1, B-2610 Antwerp, Belgium.
| | - Quinten Beirinckx
- University of Antwerp, Department of Physics, Universiteitsplein 1, B-2610 Antwerp, Belgium.
| | - Kevin Nys
- University of Antwerp, Department of Physics, Universiteitsplein 1, B-2610 Antwerp, Belgium.
| | - María Florencia Mangiameli
- Universidad National de Rosario, Departamento de Químico Física - Área Química General, Facultad de Ciencias Bioquímicas y Farmacéuticas, IQUIR-CONICET, Suipacha 531, Rosario, Santa Fe, Argentina.
| | - Bert Cuypers
- University of Antwerp, Department of Physics, Universiteitsplein 1, B-2610 Antwerp, Belgium.
| | - Freddy Callens
- Ghent University, Dept. of Solid State Sciences, Krijgslaan 281-S1, B-9000 Ghent, Belgium.
| | - Henk Vrielinck
- Ghent University, Dept. of Solid State Sciences, Krijgslaan 281-S1, B-9000 Ghent, Belgium.
| | - Juan Carlos González
- University of Antwerp, Department of Physics, Universiteitsplein 1, B-2610 Antwerp, Belgium; Universidad National de Rosario, Departamento de Químico Física - Área Química General, Facultad de Ciencias Bioquímicas y Farmacéuticas, IQUIR-CONICET, Suipacha 531, Rosario, Santa Fe, Argentina.
| |
Collapse
|
42
|
Luczak MW, Green SE, Zhitkovich A. Different ATM Signaling in Response to Chromium(VI) Metabolism via Ascorbate and Nonascorbate Reduction: Implications for in Vitro Models and Toxicogenomics. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:61-6. [PMID: 25977998 PMCID: PMC4710604 DOI: 10.1289/ehp.1409434] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 05/13/2015] [Indexed: 06/04/2023]
Abstract
BACKGROUND Carcinogenic hexavalent chromium [Cr(VI)] requires cellular reduction to generate DNA damage. Metabolism of Cr(VI) by its principal reducer ascorbate (Asc) lacks a Cr(V) intermediate, which is abundant in reactions with a minor reducing agent, glutathione. Cultured cells are widely used in mechanistic studies of Cr(VI) toxicity; however, they typically contain < 1% of normal Asc levels. Asc deficiency is also expected to diminish protection against reactive oxygen species. OBJECTIVES We assessed how the presence of Asc in cells affects their stress signaling and survival responses to chromate. METHODS We investigated the effects of Asc restoration in human lung H460 cells and normal human lung fibroblasts on the activation and functional role of ATM kinase, which controls DNA damage responses involving several hundreds of proteins. RESULTS Treatment of standard cultures with Cr(VI) strongly activated ATM, as indicated by its automodification at Ser1981 and by phosphorylation of checkpoint kinase 2 (CHK2) and chromatin/transcription regulator KRAB-associated protein 1 (KAP1). Confirming the importance of activated ATM, its inhibition impaired replication recovery and clonogenic survival. In contrast, fully Asc-restored cells lacked ATM activation by Cr(VI), and ATM silencing produced no significant effects on p53 stabilization, apoptosis, replication recovery, or clonogenic survival. Dose dependence studies found a close correlation between ATM activation and the extent of Cr(VI) reduction by glutathione. CONCLUSIONS Asc restoration in cultured cells dramatically altered their stress responses to Cr(VI) by preventing activation of the oxidant-sensitive ATM network. We suggest that toxicogenomic and other cell response-based approaches likely underestimate Cr(VI) genotoxicity when standard ATM-activating carcinogens are used as references. CITATION Luczak MW, Green SE, Zhitkovich A. 2016. Different ATM signaling in response to chromium(VI) metabolism via ascorbate and nonascorbate reduction: implications for in vitro models and toxicogenomics. Environ Health Perspect 124:61-66; http://dx.doi.org/10.1289/ehp.1409434.
Collapse
Affiliation(s)
- Michal W. Luczak
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | - Samantha E. Green
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | - Anatoly Zhitkovich
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
43
|
Thompson CM, Wolf JC, Elbekai RH, Paranjpe MG, Seiter JM, Chappell MA, Tappero RV, Suh M, Proctor DM, Bichteler A, Haws LC, Harris MA. Duodenal crypt health following exposure to Cr(VI): Micronucleus scoring, γ-H2AX immunostaining, and synchrotron X-ray fluorescence microscopy. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2015; 789-790:61-6. [PMID: 26232259 DOI: 10.1016/j.mrgentox.2015.05.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 05/07/2015] [Accepted: 05/10/2015] [Indexed: 12/18/2022]
Abstract
Lifetime exposure to high concentrations of hexavalent chromium [Cr(VI)] in drinking water results in intestinal damage and an increase in duodenal tumors in B6C3F1 mice. To assess whether these tumors could be the result of a direct mutagenic or genotoxic mode of action, we conducted a GLP-compliant 7-day drinking water study to assess crypt health along the entire length of the duodenum. Mice were exposed to water (vehicle control), 1.4, 21, or 180 ppm Cr(VI) via drinking water for 7 consecutive days. Crypt enterocytes in Swiss roll sections were scored as normal, mitotic, apoptotic, karyorrhectic, or as having micronuclei. A single oral gavage of 50mg/kg cyclophosphamide served as a positive control for micronucleus induction. Exposure to 21 and 180 ppm Cr(VI) significantly increased the number of crypt enterocytes. Micronuclei and γ-H2AX immunostaining were not elevated in the crypts of Cr(VI)-treated mice. In contrast, treatment with cyclophosphamide significantly increased numbers of crypt micronuclei and qualitatively increased γ-H2AX immunostaining. Synchrotron-based X-ray fluorescence (XRF) microscopy revealed the presence of strong Cr fluorescence in duodenal villi, but negligible Cr fluorescence in the crypt compartment. Together, these data indicate that Cr(VI) does not adversely effect the crypt compartment where intestinal stem cells reside, and provide additional evidence that the mode of action for Cr(VI)-induced intestinal cancer in B6C3F1 mice involves chronic villous wounding resulting in compensatory crypt enterocyte hyperplasia.
Collapse
Affiliation(s)
| | - Jeffrey C Wolf
- Experimental Pathology Laboratories, Sterling, VA 20166, USA.
| | | | | | - Jennifer M Seiter
- U.S. Army Engineer Research and Development Center, Vicksburg, MS 39180, USA.
| | - Mark A Chappell
- U.S. Army Engineer Research and Development Center, Vicksburg, MS 39180, USA.
| | - Ryan V Tappero
- Photon Sciences Department, Brookhaven National Laboratory, Upton, NY 11973, USA.
| | - Mina Suh
- ToxStrategies, Inc., Mission Viejo, CA 92692, USA.
| | | | | | | | | |
Collapse
|