1
|
Araujo ANM, Leroux IN, Furtado DZS, Ferreira APSDS, Batista BL, Silva HDT, Handakas E, Assunção NA, Olympio KPK. Integration of proteomic and metabolomic analyses: New insights for mapping informal workers exposed to potentially toxic elements. Front Public Health 2023; 10:899638. [PMID: 36761330 PMCID: PMC9905639 DOI: 10.3389/fpubh.2022.899638] [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: 03/18/2022] [Accepted: 12/29/2022] [Indexed: 01/26/2023] Open
Abstract
Occupational exposure to potentially toxic elements (PTEs) is a concerning reality of informal workers engaged in the jewelry production chain that can lead to adverse health effects. In this study, untargeted proteomic and metabolomic analyses were employed to assess the impact of these exposures on informal workers' exposome in Limeira city, São Paulo state, Brazil. PTE levels (Cr, Mn, Ni, Cu, Zn, As, Cd, Sn, Sb, Hg, and Pb) were determined in blood, proteomic analyses were performed for saliva samples (n = 26), and metabolomic analyses in plasma (n = 145) using ultra-high performance liquid chromatography (UHPLC) coupled with quadrupole-time-of-flight (Q-TOF) mass spectrometry. Blood PTE levels of workers, controls, and their family members were determined by inductively coupled plasma-mass spectrometry (ICP-MS). High concentration levels of Sn and Cu were detected in welders' blood (p < 0.001). Statistical analyses were performed using MetaboAnalyst 4.0. The results showed that 26 proteins were upregulated, and 14 proteins downregulated on the welder group, and thirty of these proteins were also correlated with blood Pb, Cu, Sb, and Sn blood levels in the welder group (p < 0.05). Using gene ontology analysis of these 40 proteins revealed the biological processes related to the upregulated proteins were translational initiation, SRP-dependent co-translational protein targeting to membrane, and viral transcription. A Metabolome-Wide Association Study (MWAS) was performed to search for associations between blood metabolites and exposure groups. A pathway enrichment analysis of significant features from the MWAS was then conducted with Mummichog. A total of 73 metabolomic compounds and 40 proteins up or down-regulated in welders were used to perform a multi-omics analysis, disclosing seven metabolic pathways potentially disturbed by the informal work: valine leucine and isoleucine biosynthesis, valine leucine and isoleucine degradation, arginine and proline metabolism, ABC transporters, central carbon metabolism in cancer, arachidonic acid metabolism and cysteine and methionine metabolism. The majority of the proteins found to be statistically up or downregulated in welders also correlated with at least one blood PTE level, providing insights into the biological responses to PTE exposures in the informal work exposure scenario. These findings shed new light on the effects of occupational activity on workers' exposome, underscoring the harmful effects of PTE.
Collapse
Affiliation(s)
- Alda Neis Miranda Araujo
- Graduate Program in Translational Medicine, Paulista School of Medicine, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Isabelle Nogueira Leroux
- School of Public Health, Department of Environmental Health, University of São Paulo, São Paulo, Brazil
| | - Danielle Zildeana Sousa Furtado
- Department of Chemistry, Institute of Environmental, Chemical, and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, São Paulo, Brazil,Technology School of Teresina, Teresina, Piauí, Brazil
| | | | - Bruno Lemos Batista
- Center for Natural and Human Sciences, Federal University of ABC, São Paulo, Brazil
| | - Heron Dominguez Torres Silva
- Department of Chemistry, Institute of Environmental, Chemical, and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, São Paulo, Brazil
| | - Evangelos Handakas
- Department of Medicine, Computation and Medicine, Imperial College London, London, United Kingdom
| | - Nilson Antônio Assunção
- Department of Chemistry, Institute of Environmental, Chemical, and Pharmaceutical Sciences, Federal University of São Paulo, Diadema, São Paulo, Brazil,Nilson Antônio Assunção ✉
| | - Kelly Polido Kaneshiro Olympio
- School of Public Health, Department of Environmental Health, University of São Paulo, São Paulo, Brazil,*Correspondence: Kelly Polido Kaneshiro Olympio ✉
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
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
|
5
|
Ding SZ, Yang YX, Li XL, Michelli-Rivera A, Han SY, Wang L, Pratheeshkumar P, Wang X, Lu J, Yin YQ, Budhraja A, Hitron AJ. Epithelial-mesenchymal transition during oncogenic transformation induced by hexavalent chromium involves reactive oxygen species-dependent mechanism in lung epithelial cells. Toxicol Appl Pharmacol 2013; 269:61-71. [PMID: 23518002 DOI: 10.1016/j.taap.2013.03.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Revised: 03/07/2013] [Accepted: 03/08/2013] [Indexed: 12/18/2022]
Abstract
Hexavalent chromium [Cr(VI)] is an important human carcinogen associated with pulmonary diseases and lung cancer. Exposure to Cr(VI) induces DNA damage, cell morphological change and malignant transformation in human lung epithelial cells. Despite extensive studies, the molecular mechanisms remain elusive, it is also not known if Cr(VI)-induced transformation might accompany with invasive properties to facilitate metastasis. We aimed to study Cr(VI)-induced epithelial-mesenchymal transition (EMT) and invasion during oncogenic transformation in lung epithelial cells. The results showed that Cr(VI) at low doses represses E-cadherin mRNA and protein expression, enhances mesenchymal marker vimentin expression and transforms the epithelial cell into fibroblastoid morphology. Cr(VI) also increases cell invasion and promotes colony formation. Further studies indicated that Cr(VI) uses multiple mechanisms to repress E-cadherin expression, including activation of E-cadherin repressors such as Slug, ZEB1, KLF8 and enhancement the binding of HDAC1 in E-cadherin gene promoter, but DNA methylation is not responsible for the loss of E-cadherin. Catalase reduces Cr(VI)-induced E-cadherin and vimentin protein expression, attenuates cell invasion in matrigel and colony formation on soft agar. These results demonstrate that exposure to a common human carcinogen, Cr(VI), induces EMT and invasion during oncogenic transformation in lung epithelial cells and implicate in cancer metastasis and prevention.
Collapse
Affiliation(s)
- Song-Ze Ding
- Department of Internal Medicine, Henan Provincial People's Hospital, Zhengzhou University, Wei-Wu Road, Zhengzhou, Henan 450000, PR China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Fan Y, Ovesen JL, Puga A. Long-term exposure to hexavalent chromium inhibits expression of tumor suppressor genes in cultured cells and in mice. J Trace Elem Med Biol 2012; 26:188-91. [PMID: 22613061 PMCID: PMC3380135 DOI: 10.1016/j.jtemb.2012.04.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We have used mouse hepatoma cells in culture to study acute, short-term high-dose effects of hexavalent chromium on gene regulation directed by the polycyclic aromatic hydrocarbon benzo[a]pyrene (BaP). We find that the mixture engages three major signaling pathways: (i) activation of detoxification genes; (ii) induction of signal transduction effectors; and (iii) epigenetic modification of chromatin marks. Preliminary results in mice exposed to mixtures of low doses of Cr(VI) plus BaP indicate that all three pathways are likely to be engaged also in long-term effects resulting from exposure to environmentally relevant doses of the mixture that inhibit the expression of tumor suppressor genes. Given the toxicity and carcinogenicity of these mixtures, we expect that a two-way analytical approach, from cells in culture to biological effects in vivo and vice versa, will provide a better understanding of the molecular mechanisms responsible for the biological effects of mixtures. By focusing both the in vivo and the in vitro work into long-term, low-dose, environmentally relevant exposures, we expect to develop much needed information pertinent to the type of diseases found in human populations exposed to mixtures of environmental toxicants.
Collapse
Affiliation(s)
- Yunxia Fan
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati, College of Medicine, 3223 Eden Ave. Cincinnati, OH 45267
| | - Jerald L. Ovesen
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati, College of Medicine, 3223 Eden Ave. Cincinnati, OH 45267
| | - Alvaro Puga
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati, College of Medicine, 3223 Eden Ave. Cincinnati, OH 45267
| |
Collapse
|
7
|
Sundaram B, Singhal K, Sandhir R. Ameliorating effect of chromium administration on hepatic glucose metabolism in streptozotocin-induced experimental diabetes. Biofactors 2012; 38:59-68. [PMID: 22287284 DOI: 10.1002/biof.194] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2011] [Accepted: 12/07/2011] [Indexed: 02/02/2023]
Abstract
Chromium has been recognized as an essential trace element that plays an important role in carbohydrate metabolism. However, the molecular mechanisms involved in its action are not clear. This study was undertaken to understand the mechanism of chromium action in experimental diabetes. Streptozotocin-induced diabetic animals were administered chromium as chromium picolinate (CrP) at a daily dose of 1 mg/kg body weight for a period of 4 weeks. It was observed that chromium complexed with picolinate was effective in lowering plasma glucose levels as well as was able to alleviate polyphagia, polydipsia, and weight loss in diabetic animals. Administration of chromium was also found to normalize glycogen content in liver of diabetic animals to near control levels. The reduction in plasma glucose levels by chromium was accompanied by increase in activity of glycolytic enzymes (e.g., glucokinase, phosphofructokinase, and pyruvate kinase) and by suppression in activity of gluconeogenic enzymes (e.g., glucose-6-phosphatase and phosphoenolpyruvate carboxykinase) in liver. Hepatic glucose uptake was found to be increased by chromium supplementation as demonstrated by decrease in Km and increase in Vmax values in diabetic animals. Chromium levels were lower in the liver of diabetic rats when compared with that of control rats. A negative correlation was observed between plasma glucose and chromium concentration in patients with diabetes. The data suggests that chromium supplementation as CrP is beneficial in correcting hyperglycemia, implying that the modulation of the glucose metabolism by chromium may be therapeutically beneficial in the treatment of diabetes.
Collapse
|
8
|
Marketon JIW, Sternberg EM. The glucocorticoid receptor: a revisited target for toxins. Toxins (Basel) 2010; 2:1357-80. [PMID: 22069642 PMCID: PMC3153245 DOI: 10.3390/toxins2061357] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2010] [Revised: 05/28/2010] [Accepted: 06/07/2010] [Indexed: 12/15/2022] Open
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis activation and glucocorticoid responses are critical for survival from a number of bacterial, viral and toxic insults, demonstrated by the fact that removal of the HPA axis or GR blockade enhances mortality rates. Replacement with synthetic glucocorticoids reverses these effects by providing protection against lethal effects. Glucocorticoid resistance/insensitivity is a common problem in the treatment of many diseases. Much research has focused on the molecular mechanism behind this resistance, but an area that has been neglected is the role of infectious agents and toxins. We have recently shown that the anthrax lethal toxin is able to repress glucocorticoid receptor function. Data suggesting that the glucocorticoid receptor may be a target for a variety of toxins is reviewed here. These studies have important implications for glucocorticoid therapy.
Collapse
Affiliation(s)
- Jeanette I. Webster Marketon
- Department of Internal Medicine, Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, 201 DHLRI, 473 W. 12th Avenue, Columbus, OH 43210, USA
- Institute for Behavioral Medicine Research, The Ohio State University Medical Center, 460 Medical Center Drive, Columbus, OH 43210, USA
- Author to whom correspondence should be addressed; ; Tel.: +1-614-293-3496; Fax: +1-614-366-2074
| | - Esther M. Sternberg
- Department of Health and Human Services, Section on Neuroendocrine Immunology and Behavior, National Institute of Mental Health, National Institutes of Health, 5625 Fishers Lane, Rm. 4N13 (MSC 9401), Bethesda, MD 20892-9401, USA;
| |
Collapse
|
9
|
Macfie A, Hagan E, Zhitkovich A. Mechanism of DNA-protein cross-linking by chromium. Chem Res Toxicol 2010; 23:341-7. [PMID: 19877617 DOI: 10.1021/tx9003402] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hexavalent chromium is a known inducer of DNA-protein cross-links (DPCs) that contribute to repression of inducible genes and genotoxicity of this metal. Lymphocytic DPCs have also shown potential utility as biomarkers of human exposure to Cr(VI). Here, we examined the mechanism of DPC formation by Cr(VI) and the impact of its main cellular reducers. In vitro reactions of Cr(VI) with one-electron reducing thiols (glutathione and cysteine) or two-electron donating ascorbate were all efficient at DPC production, indicating a dispensable role of Cr(V). No Cr(VI) reducer was able to generate DPC in the presence of Cr(III)-chelating EDTA or phosphate. A critical role of Cr(III) in DNA-protein linkages was further confirmed by dissociation of Cr(VI)-induced DPC by phosphate. EDTA was very inefficient in DPC dissociation, indicating its poor suitability for testing of Cr(III)-mediated bridging and reversal of complex DPC. Reactions containing only one Cr-modified component (protein or DNA) showed that Cr(III)-DNA adduction was the initial step in DPC formation. Cross-linking proceeded slowly after the rapid formation of Cr-DNA adducts, indicating that protein conjugation was the rate-limiting step in DPC generation. Experiments with depletion of glutathione and restoration of ascorbate levels in human lung A549 cells showed that high cellular reducing capacity promotes DPC yield. Overall, our data provide evidence for a three-step cross-linking mechanism involving (i) reduction of Cr(VI) to Cr(III), (ii) Cr(III)-DNA binding, and (iii) protein capture by DNA-bound Cr(III) generating protein-Cr(III)-DNA cross-links.
Collapse
Affiliation(s)
- Andrea Macfie
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship Street, Providence, Rhode Island 02912, USA
| | | | | |
Collapse
|
10
|
Schnekenburger M, Talaska G, Puga A. Chromium cross-links histone deacetylase 1-DNA methyltransferase 1 complexes to chromatin, inhibiting histone-remodeling marks critical for transcriptional activation. Mol Cell Biol 2007; 27:7089-101. [PMID: 17682057 PMCID: PMC2168892 DOI: 10.1128/mcb.00838-07] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Transcriptional regulation of gene expression requires posttranslational modification of histone proteins, which, in concert with chromatin-remodeling factors, modulate chromatin structure. Exposure to environmental agents may interfere with specific histone modifications and derail normal patterns of gene expression. To test this hypothesis, we coexposed cells to binary mixtures of benzo[a]pyrene (B[a]P), an environmental procarcinogen that activates Cyp1a1 transcriptional responses mediated by the aryl hydrocarbon receptor (AHR), and chromium, a carcinogenic heavy metal that represses B[a]P-inducible AHR-mediated gene expression. We show that chromium cross-links histone deacetylase 1-DNA methyltransferase 1 (HDAC1-DNMT1) complexes to Cyp1a1 promoter chromatin and inhibits histone marks induced by AHR-mediated gene transactivation, including phosphorylation of histone H3 Ser-10, trimethylation of H3 Lys-4, and various acetylation marks in histones H3 and H4. These changes inhibit RNA polymerase II recruitment without affecting the kinetics of AHR DNA binding. HDAC1 and DNMT1 inhibitors or depletion of HDAC1 or DNMT1 with siRNAs blocks chromium-induced transcriptional repression by decreasing the interaction of these proteins with the Cyp1a1 promoter and allowing histone acetylation to proceed. By inhibiting Cyp1a1 expression, chromium stimulates the formation of B[a]P DNA adducts. Epigenetic modification of gene expression patterns may be a key element of the developmental and carcinogenic outcomes of exposure to chromium and to other environmental agents.
Collapse
Affiliation(s)
- Michael Schnekenburger
- Department of Environmental Health, University of Cincinnati Medical Center, P.O. Box 670056, Cincinnati, OH 45267-0056, USA
| | | | | |
Collapse
|
11
|
Wei YD, Tepperman K, Huang MY, Sartor MA, Puga A. Chromium inhibits transcription from polycyclic aromatic hydrocarbon-inducible promoters by blocking the release of histone deacetylase and preventing the binding of p300 to chromatin. J Biol Chem 2003; 279:4110-9. [PMID: 14625279 DOI: 10.1074/jbc.m310800200] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Co-contamination with complex mixtures of carcinogenic metals, such as chromium, and polycyclic aromatic hydrocarbons is a common environmental problem with multiple biological consequences. Chromium exposure alters inducible gene expression, forms chromium-DNA adducts and chromium-DNA cross-links, and disrupts transcriptional activator-co-activator complexes. We have shown previously that exposure of mouse hepatoma Hepa-1 cells to chromate inhibits the induction of the Cyp1a1 and Nqo1 genes by dioxin. Here we have tested the hypothesis that chromium blocks gene expression by interfering with the assembly of productive transcriptional complexes at the promoter of inducible genes. To this end, we have studied the effects of chromium on the expression of genes induced by benzo[a]pyrene (B[a]P), another aryl hydrocarbon receptor agonist, and characterized the disruption of Cyp1a1 transcriptional induction by chromium. Gene expression profiling by using high density microarray analysis revealed that the inhibitory effect of chromium on B[a]P-dependent gene induction was generalized, affecting the induction of over 50 different genes involved in a variety of signaling transduction pathways. The inhibitory effect of chromium on Cyp1a1 transcription was found to depend on the presence of promoter-proximal sequences and not on the cis-acting enhancer sequences that bind the aryl hydrocarbon receptor-aryl hydrocarbon receptor nuclear translocator complex. By using transient reporter assays and chromatin immunoprecipitation analyses, we found that chromium prevented the B[a]P-dependent release of HDAC-1 from Cyp1a1 chromatin and blocked p300 recruitment. These results provide a mechanistic explanation for the observation that chromium inhibits inducible but not constitutive gene expression.
Collapse
Affiliation(s)
- Yu-Dan Wei
- Center for Environmental Genetics and Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio 45220-0056, USA
| | | | | | | | | |
Collapse
|
12
|
Klein CB, Su L, Bowser D, Leszczynska J. Chromate-induced epimutations in mammalian cells. ENVIRONMENTAL HEALTH PERSPECTIVES 2002; 110 Suppl 5:739-43. [PMID: 12426123 PMCID: PMC1241236 DOI: 10.1289/ehp.02110s5739] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Epigenetic gene silencing by aberrant DNA methylation of gene promoter regions is a nonmutagenic but heritable epigenetic mechanism that may mistakenly cause the silencing of important cancer-related tumor suppressor genes. Using a transgenic, V79-derived, mammalian cell line (G12) that contains a bacterial gpt reporter gene in its DNA, we can study carcinogen-induced gene inactivation by mutagenic as well as epigenetic DNA methylation mechanisms. Whereas numerous carcinogens have previously been shown to be mutagenic in these cells, a few carcinogens, including nickel, diethylstilbestrol, and X-rays, are also capable of silencing the G12 cell gpt transgene by aberrant DNA methylation. Here we report for the first time that carcinogenic potassium chromate salts can also induce aberrant DNA methylation in this system. In contrast insoluble barium chromate produced significant level of mutations in these cells but did not cause DNA methylation changes associated with transgene expression.
Collapse
Affiliation(s)
- Catherine B Klein
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA.
| | | | | | | |
Collapse
|
13
|
Maitra R, Halpin PA, Karlson KH, Page RL, Paik DY, Leavitt MO, Moyer BD, Stanton BA, Hamilton JW. Differential effects of mitomycin C and doxorubicin on P-glycoprotein expression. Biochem J 2001; 355:617-24. [PMID: 11311122 PMCID: PMC1221775 DOI: 10.1042/bj3550617] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Previous studies have demonstrated that mitomycin C (MMC) and other DNA cross-linking agents can suppress MDR1 (multidrug resistance 1) gene expression and subsequent functional P-glycoprotein (Pgp) expression, whereas doxorubicin and other anthracyclines increase MDR1 gene expression. In the present study, with stably transfected Madin-Darby canine kidney C7 epithelial cells expressing a human Pgp tagged with green fluorescent protein under the proximal human MDR1 gene promoter, we demonstrated that MMC and doxorubicin have differential effects on Pgp expression and function. Doxorubicin caused a progressive increase in the cell-surface expression of Pgp and function. In contrast, MMC initially increased plasma membrane expression and function at a time when total cellular Pgp was constant and Pgp mRNA expression had been shown to be suppressed. This was followed by a rapid and sustained decrease in cell-surface expression at later times, presumably as a consequence of the initial decrease in mRNA expression. These studies imply that there are at least two independent chemosensitive steps that can alter Pgp biogenesis: one at the level of mRNA transcription and the other at the level of Pgp trafficking. Understanding the combined consequences of these two mechanisms might lead to novel chemotherapeutic approaches to overcoming drug resistance in human cancers by altering either Pgp mRNA expression or trafficking to the membrane.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis
- ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- Alkylating Agents/pharmacology
- Animals
- Antibiotics, Antineoplastic/pharmacology
- Biological Transport/drug effects
- Doxorubicin/pharmacology
- Gene Expression/drug effects
- Humans
- Mitomycin/pharmacology
- RNA, Messenger/biosynthesis
- RNA, Messenger/drug effects
- Rats
- Transcription, Genetic/drug effects
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- R Maitra
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, NH 03755-3835, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Maier A, Dalton TP, Puga A. Disruption of dioxin-inducible phase I and phase II gene expression patterns by cadmium, chromium, and arsenic. Mol Carcinog 2000. [DOI: 10.1002/1098-2744(200008)28:4<225::aid-mc5>3.0.co;2-o] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
15
|
Kaltreider RC, Pesce CA, Ihnat MA, Lariviere JP, Hamilton JW. Differential effects of arsenic(III) and chromium(VI) on nuclear transcription factor binding. Mol Carcinog 1999. [DOI: 10.1002/(sici)1098-2744(199907)25:3<219::aid-mc8>3.0.co;2-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
16
|
Cohen MD, Zelikoff JT, Chen LC, Schlesinger RB. Immunotoxicologic effects of inhaled chromium: role of particle solubility and co-exposure to ozone. Toxicol Appl Pharmacol 1998; 152:30-40. [PMID: 9772197 DOI: 10.1006/taap.1998.8502] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Soluble and insoluble hexavalent chromium (Cr6+) agents are concomitantly released with ozone (O3) during welding. Although pulmonary/immunologic implications from exposure to each agent individually have been investigated, the effects from simultaneous exposure, as occurs under actual working conditions, are unclear. To investigate immunomodulatory effects of inhaled Cr6+, F-344 rats were exposed for 5 h/day, 5 days/week for 2 or 4 weeks to atmospheres containing soluble potassium chromate (K2CrO4) or insoluble barium chromate (BaCrO4), each alone at 360 micrograms Cr/m3 or in combination with 0.3 ppm O3. One day after the final exposure, rats were euthanized, their lungs were lavaged, and pulmonary macrophages (PAM) were recovered for assessment of basal and inducible functions. Rats inhaling K2CrO4-containing atmospheres had greater levels of total recoverable cells, neutrophils, and monocytes in bronchopulmonary lavage compared to rats exposed to insoluble Cr6+ atmospheres, O3 alone, or air; these rats also had a reduced percentage of PAM, although total PAM levels remained unaffected. Although Cr exposure-related changes in PAM functionality were evident, any dependence upon Cr solubility was variable. K2CrO4-containing atmospheres modulated PAM-inducible interleukins-1 and -6, and tumor necrosis factor-alpha production to a greater degree than those containing BaCrO4. Conversely, BaCrO4-containing atmospheres affected PAM basal nitric oxide production and interferon-gamma-primed/zymosan-stimulated reactive oxygen intermediate production to a greater extent than did those containing K2CrO4. In none of the PAM assays did co-inhalation of O3 result in a modulation of the effects obtained with either Cr6+ compound itself. The results indicate that, while immunomodulatory effects of inhaled Cr6+ upon PAM are related to particle solubility, the co-inhalation of O3 apparently does not cause further modifications of the metal-induced effects.
Collapse
Affiliation(s)
- M D Cohen
- Department of Environmental Medicine, New York University Medical Center, Tuxedo, New York 10987, USA
| | | | | | | |
Collapse
|
17
|
Hamilton JW, Kaltreider RC, Bajenova OV, Ihnat MA, McCaffrey J, Turpie BW, Rowell EE, Oh J, Nemeth MJ, Pesce CA, Lariviere JP. Molecular basis for effects of carcinogenic heavy metals on inducible gene expression. ENVIRONMENTAL HEALTH PERSPECTIVES 1998; 106 Suppl 4:1005-15. [PMID: 9703486 PMCID: PMC1533345 DOI: 10.1289/ehp.98106s41005] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Certain forms of the heavy metals arsenic and chromium are considered human carcinogens, although they are believed to act through very different mechanisms. Chromium(VI) is believed to act as a classic and mutagenic agent, and DNA/chromatin appears to be the principal target for its effects. In contrast, arsenic(III) is considered nongenotoxic, but is able to target specific cellular proteins, principally through sulfhydryl interactions. We had previously shown that various genotoxic chemical carcinogens, including chromium (VI), preferentially altered expression of several inducible genes but had little or no effect on constitutive gene expression. We were therefore interested in whether these carcinogenic heavy metals might target specific but distinct sites within cells, leading to alterations in gene expression that might contribute to the carcinogenic process. Arsenic(III) and chromium(VI) each significantly altered both basal and hormone-inducible expression of a model inducible gene, phosphoenolpyruvate carboxykinase (PEPCK), at nonovertly toxic doses in the chick embryo in vivo and rat hepatoma H411E cells in culture. We have recently developed two parallel cell culture approaches for examining the molecular basis for these effects. First, we are examining the effects of heavy metals on expression and activation of specific transcription factors known to be involved in regulation of susceptible inducible genes, and have recently observed significant but different effects of arsenic(III) and chromium(VI) on nuclear transcription factor binding. Second, we have developed cell lines with stably integrated PEPCK promoter-luciferase reporter gene constructs to examine effects of heavy metals on promoter function, and have also recently seen profound effects induced by both chromium(VI) and arsenic(III) in this system. These model systems should enable us to be able to identify the critical cis (DNA) and trans (protein) cellular targets of heavy metal exposure leading to alterations in expression of specific susceptible genes. It is anticipated that such information will provide valuable insight into the mechanistic basis for these effects as well as provide sensitive molecular biomarkers for evaluating human exposure.
Collapse
Affiliation(s)
- J W Hamilton
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, NH 03755-3835, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Warren AJ, Hamilton JW. Synthesis and structural characterization of the N2G-mitomycin C-N2G interstrand cross-link in a model synthetic 23 base pair oligonucleotide DNA duplex. Chem Res Toxicol 1996; 9:1063-71. [PMID: 8902260 DOI: 10.1021/tx960070c] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Mitomycin C (MMC) is a genotoxic cancer chemotherapeutic agent that reacts principally at the N2 position of guanine to form one of two predominant monoadducts, or a G-G interstrand cross-link at CpG sites, or a G-G intrastrand cross-link at GpG sites. Previous studies of MMC adduction have principally used very short duplex oligonucleotides (5-15 bp) or very long native duplex DNAs. We examined the formation and structural features of the MMC CpG interstrand cross-link on a model 23 bp synthetic oligonucleotide duplex having the (upper strand) sequence 5'-ATAAATACGTATTTATTTATAAA-3'. MMC was reacted with the duplex oligonucleotide in the presence of sodium dithionite at ratios of 6 mM dithionite: 1.5 mM MMC:0.03 mM duplex. The yield of cross-link in the reaction was determined to be approximately 4.8% by denaturing gel electrophoresis, which represented approximately 75% of the total bound MMC. The cross-linked DNA was isolated to greater than 97% purity in a single step by high temperature size exclusion column chromatography. Characterization of the purified product confirmed that the complex contained exclusively the N2G-MMC-N2G cross-link at the single central CpG site. CD spectroscopy demonstrated a negative band at approximately 290-320 nm which has previously been shown to be characteristic of the MMC cross-link. The relative intensity of this band compared to those reported for shorter duplexes suggested that the majority of the duplex is in a normal B-DNA helical configuration. Base-specific chemical footprinting techniques also indicated that there were subtle but distinct structural perturbations principally within the central four to six base pairs containing and adjacent to the cross-link.
Collapse
Affiliation(s)
- A J Warren
- Department of Chemistry, Dartmouth College, Hanover, New Hampshire 03755-3564, USA
| | | |
Collapse
|
19
|
Caron RM, Hamilton JW. Preferential effects of the chemotherapeutic DNA crosslinking agent mitomycin C on inducible gene expression in vivo. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 1995; 25:4-11. [PMID: 7875125 DOI: 10.1002/em.2850250103] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
The immediate effects of a single dose of the chemotherapeutic DNA crosslinking agent, mitomycin C (MMC), on the expression of several constitutive and drug-inducible genes were examined in a simple in vivo system, the 14 day chick embryo. We observed no effect of MMC on the steady-state mRNA expression of the constitutively expressed beta-actin, transferrin, or albumin genes. In contrast, MMC treatment significantly altered both the basal and drug-inducible mRNA expression of two glutethimide-inducible genes, 5-aminolevulinic acid (ALA) synthase and cytochrome P450 CYP2H1. The basal expression of these genes was transiently but significantly increased over a 24 hr period following a single dose of MMC. Conversely, MMC significantly suppressed the glutethimide-inducible expression of these genes when administered 1 to 24 hr prior to the inducing drug. The effects of MMC on both basal and drug-inducible ALA synthase and CYP2H1 mRNA expression were principally a result of changes in the transcription rates of these genes. In contrast, MMC treatment had little or no effect on glutethimide-induced expression of ALA synthase or CYP2H1 when administered 1 hr after the inducing drug, suggesting that a very early event in the induction process represents the target for these MMC effects. Covalent binding studies demonstrated that the effects of MMC on gene expression were closely correlated temporally with formation of [3H]-porfiromycin-DNA adducts. These results support the hypothesis that genotoxic chemicals specifically target their effects to inducible genes in vivo.
Collapse
Affiliation(s)
- R M Caron
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, NH 03755-3835
| | | |
Collapse
|