Elsevier

Seminars in Cancer Biology

Volume 76, November 2021, Pages 54-60
Seminars in Cancer Biology

Hexavalent chromium disrupts chromatin architecture

https://doi.org/10.1016/j.semcancer.2021.07.009Get rights and content

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.

Introduction

Chromium is an abundant transition metal that is most commonly found in the environment as one of three stable valences, as either trivalent (Cr(III)), hexavalent (Cr(VI)) or metallic (Cr(0). While Cr(III) is the most common form that occurs naturally, the origins of Cr(VI) compounds can largely be attributed to anthropogenic sources due to the industrial use of chromium products in stainless-steel production, electroplating, leather tanning, textile and pigment production, among others [1]. Trivalent chromium can be harmful in acute exposures, however the hexavalent form is considered to be substantially more toxic on account of its increased ability to cross cellular membranes. On the other hand, Cr(III) relies on passive diffusion to permeate the cell membrane, substantially limiting its accumulation within cells. While Cr(VI) is largely reduced in the extracellular environment to Cr(III), molecules that escape the initial detoxification process are rapidly facilitated into the cell through nonspecific sulfate/phosphate anion transporters and subsequently reduced to Cr(III) by ascorbate, glutathione, and to a lesser extent cysteine, through one- or two- step reductions (depending on the reducing agent) [[2], [3], [4], [5], [6]].

Consequently, studies investigating the toxicity of chromium largely focus on the hexavalent species. Specifically, Cr(VI) has been well-established as a respiratory carcinogen through multiple epidemiological and animal studies, with an emphasis on occupational settings where chromate workers are primarily exposed through inhalation and dermal contact. However, the most common route of exposure for the general population is the ingestion of drinking water contaminated with low levels Cr(VI) which may be the result of improper disposal methods of chromium waste, pollution, and natural sources. A recent study by Tan et al. has reported that interactions between cast iron pipes and chlorine disinfectants in drinking water distribution systems can promote the formation of Cr(VI) [7]. While evidence suggesting ingestion is associated with increased cancer incidence exists, the quantification of risk in the general population is not as clear. Despite this, current models of Cr(VI) uptake and intracellular metabolism suggest that carcinogenesis is inextricably linked to the molecular properties of chromium and further understanding the mechanisms through which chromium promotes carcinogenesis is important for improving the health and safety of the population. Continued advances in our understanding of the genome highlight the critical role chromatin organization plays in the function of the cell. Based on current evidence, chromium interferes with the structure and function at multiple levels of chromatin compaction. By damaging DNA directly, through the formation of DNA adducts and protein-DNA crosslinks, Cr(VI) evokes a DNA damage response that requires extensive chromatin remodeling to accommodate the repair machinery [8,9]. At the nucleosome level, emerging evidence is showing that Cr(VI) alters chromatin accessibility by changing the nucleosomal occupancy and positional shifts [10]. Importantly, these changes occur at CTCF and AP1 binding sites, supporting a mechanism where Cr(VI) alters the DNA binding capacities of critical transcription factors, leading to an altered chromatin state. Ultimately, Cr(VI) targets active gene regulatory regions, disrupting gene transcription in biologically relevant cellular processes.

Section snippets

Cr(VI) damages DNA through direct and indirect mechanisms

The reduction of Cr(VI) in the body can be protective or toxic depending on where the reaction occurs. On one hand, reduction in the extracellular environment serves as a primary method of detoxification and largely prevents the uptake of excess Cr(III) in cells. On the other, similar processes act on Cr(VI) that is rapidly taken up inside cells and results in the production of free radicals, reactive intermediate metabolites, and accumulation of Cr(III) (Fig. 1). While Cr(VI) does not directly

Discussion and future direction

The carcinogenic mechanisms of inhaled hexavalent chromium are well documented in occupational settings with additional studies supporting evidence for carcinogenic potential through other routes of exposure, however the mechanisms promoting carcinogenesis are complex and require further elucidation. Genotoxicity appears to be an innate property of Cr(VI) and recent advances by several groups highlight the duality of a cell’s protective mechanisms in the presence of Cr(VI); the same

Funding

This research was supported by NIEHS grants R01 ES010807, and by the NIEHS Center for Environmental Genetics grant P30 ES06096. A.V.H. was supported by the NIEHS Training GrantT32 ES007250.

Declaration of Competing Interest

The authors declare no conflict of interest.

References (90)

  • J. Lafrance-Vanasse et al.

    Envisioning the dynamics and flexibility of Mre11-Rad50-Nbs1 complex to decipher its roles in DNA replication and repair

    Prog. Biophys. Mol. Biol.

    (2015)
  • M. Stucki et al.

    MDC1 directly binds phosphorylated histone H2AX to regulate cellular responses to DNA double-strand breaks

    Cell

    (2005)
  • S.P. Jackson et al.

    Regulation of DNA damage responses by ubiquitin and SUMO

    Mol. Cell

    (2013)
  • P. Xu

    The NuA4 core complex acetylates nucleosomal histone H4 through a double recognition mechanism

    Mol. Cell

    (2016)
  • C.L. Browning et al.

    Prolonged exposure to particulate chromate inhibits RAD51 nuclear import mediator proteins

    Toxicol. Appl. Pharmacol.

    (2017)
  • T.-C. Wang

    Oxidative DNA damage and global DNA hypomethylation are related to folate deficiency in chromate manufacturing workers

    J. Hazard. Mater.

    (2012)
  • Y. Fan et al.

    Long-term exposure to hexavalent chromium inhibits expression of tumor suppressor genes in cultured cells and in mice

    J. Trace Elem. Med. Biol.

    (2012)
  • J.L. Ovesen et al.

    Long-term exposure to low-concentrations of Cr(VI) induce DNA damage and disrupt the transcriptional response to benzo[a]pyrene

    Toxicology

    (2014)
  • Y.-D. Wei et al.

    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.

    (2004)
  • J.E. Phillips et al.

    CTCF: master Weaver of the genome

    Cell

    (2009)
  • N.S. Raja et al.

    Chromium(III) complexes inhibit transcription factors binding to DNA and associated gene expression

    Toxicology

    (2008)
  • S. Wilbur

    Toxicological Profile for Chromium

    (2012)
  • M. Valko et al.

    Metals, toxicity and oxidative stress

    Curr. Med. Chem.

    (2005)
  • G. Quievryn et al.

    Carcinogenic chromium(VI) induces cross-linking of vitamin C to DNA in vitro and in human lung A549 cells

    Biochemistry

    (2002)
  • P.H. Connett et al.

    In vitro reaction of the carcinogen chromate with cellular thiols and carboxylic acids

    J. Am. Chem. Soc.

    (2002)
  • S. DM et al.

    Reduction of chromium(VI) by ascorbate leads to chromium-DNA binding and DNA strand breaks in vitro

    Biochemistry

    (1995)
  • C. Tan et al.

    Hexavalent chromium release in drinking water distribution systems: new insights into zerovalent chromium in iron corrosion scales

    Environ. Sci. Technol.

    (2020)
  • S.S. Wise et al.

    Hexavalent chromium-induced DNA damage and repair mechanisms

    Rev. Environ. Health

    (2008)
  • Z. DeLoughery et al.

    DNA double-strand breaks by Cr(VI) are targeted to euchromatin and cause ATR-dependent phosphorylation of histone H2AX and its ubiquitination

    Toxicol. Sci.

    (2015)
  • A. VonHandorf

    Chromium disrupts chromatin organization and CTCF access to its cognate sites in promoters of differentially expressed genes

    Epigenetics

    (2018)
  • G. Waris et al.

    Reactive oxygen species: role in the development of cancer and various chronic conditions

    J. Carcinog.

    (2006)
  • X. Shi et al.

    ESR spin trapping detection of hydroxyl radicals in the reactions of Cr(V) complexes with hydrogen peroxide

    Free Radic. Res. Commun.

    (1990)
  • J. Cadet et al.

    DNA base damage by reactive oxygen species, oxidizing agents, and UV radiation

    Cold Spring Harb. Perspect. Biol.

    (2013)
  • F. Gorini

    The genomic landscape of 8-oxodG reveals enrichment at specific inherently fragile promoters

    Nucleic Acids Res.

    (2020)
  • J. Šebera

    The mechanism of the glycosylase reaction with hOGG1 base-excision repair enzyme: concerted effect of Lys249 and Asp268 during excision of 8-oxoguanine

    Nucleic Acids Res.

    (2017)
  • D. Kidane et al.

    Accumulation of abasic sites induces genomic instability in normal human gastric epithelial cells during Helicobacter pylori infection

    Oncogenesis

    (2014)
  • Pda C. Fresco et al.

    The reductive conversion of chromium(VI) by ascorbate gives rise to apurinic/apyrimidinic sites in isolated DNA

    Chem. Res. Toxicol.

    (1995)
  • P.G. Slade et al.

    Guanine-specific oxidation of double-stranded DNA by Cr(VI) and ascorbic acid forms spiroiminodihydantoin and 8-Oxo-2-deoxyguanosine

    Chem. Res. Toxicol.

    (2005)
  • M. Kolbanovskiy

    The nonbulky DNA lesions spiroiminodihydantoin and 5-guanidinohydantoin significantly block human RNA polymerase II elongation in vitro

    Biochemistry

    (2017)
  • X.-H. Zhang

    Chronic occupational exposure to hexavalent chromium causes DNA damage in electroplating workers

    BMC Public Health

    (2011)
  • A. Valavanidis et al.

    8-hydroxy-2-deoxyguanosine (8-OHdG): a critical biomarker of oxidative stress and carcinogenesis

    J. Environ. Sci. Heal. Part C

    (2009)
  • A. Zhitkovich et al.

    Glutathione and free amino acids form stable complexes with DNA following exposure of intact mammalian cells to chromate

    Carcinogenesis

    (1995)
  • W. Zhou et al.

    Cr3+ binding to DNA backbone phosphate and bases: slow ligand exchange rates and metal hydrolysis

    Inorg. Chem.

    (2016)
  • S.A. Blankert et al.

    Characterization of nonmutagenic Cr(III)-DNA interactions

    Chem. Res. Toxicol.

    (2003)
  • A. Macfie et al.

    Mechanism of DNA-protein cross-linking by chromium

    Chem. Res. Toxicol.

    (2010)
  • Cited by (14)

    • Treatment of soda ash chromite ore processing residue by Waste-Molasses-Based Ball Milling: A new strategy for disposal of waste with waste

      2022, Journal of Cleaner Production
      Citation Excerpt :

      In the industrial production of chromium and chromate, a large amount of Chromium Ore Processing Residue (COPR) is produced (Antony et al., 2013). COPR contains a lot of soluble hexavalent chromium Cr(VI) with high fluidity, high oxidation state, and high toxicity, which seriously threatens human life and health (Suljevic et al., 2021; VonHandorf et al., 2021). The development of the chromate production process has gone through two stages: lime roasting and soda ash roasting (Chen et al., 2014).

    • A novel liquid crystal resonance Rayleigh scattering spectral probe for determination of trace Cr<sup>6+</sup>

      2022, Spectrochimica Acta - Part A: Molecular and Biomolecular Spectroscopy
      Citation Excerpt :

      The toxicity of Cr6+is several hundred times that of Cr3+, and the harm to the human body is much higher than that of Cr3+. Research have found that Cr6+ can directly interact with cellular proteins, DNA and other macromolecules, resulting in general damage or function changes [24]. The common analytical methods for Cr6+ are mainly spectrophotometry, atomic absorption spectrometry, electrochemical method and chromatography.

    • Selenium regulates the mitogen-activated protein kinase pathway to protect broilers from hexavalent chromium-induced kidney dysfunction and apoptosis

      2022, Ecotoxicology and Environmental Safety
      Citation Excerpt :

      Remnant Cr (VI) is transported to the nucleus, where it is reduced to Cr (III). Excessive nuclear Cr (III) binds DNA and causes DNA damage (Sawicka et al., 2021; VonHandorf et al., 2021). Moreover, the accumulation of ROS induces toxicity in various organ tissues (e.g., nephrotoxicity, hepatotoxicity, neurotoxicity, and ovarian toxicity) (Fedala et al., 2022; Thompson et al., 2020; Wise et al., 2022; Yang et al., 2022).

    View all citing articles on Scopus
    1

    These two authors contributed equally

    View full text