Elsevier

Toxicology

Volume 430, 30 January 2020, 152340
Toxicology

Review
Understanding the mechanistic insight of arsenic exposure and decoding the histone cipher

https://doi.org/10.1016/j.tox.2019.152340Get rights and content

Abstract

Background

The study of heritable epigenetic changes in arsenic exposure has intensified over the last decade. Groundwater arsenic contamination causes a great threat to humans and, to date, no accurate measure has been formulated for remediation. The fascinating possibilities of epi-therapeutics identify the need for an in-depth mechanistic understanding of the epigenetic landscape.

Objective

In this comprehensive review, we have set to analyze major studies pertaining to histone post-translational modifications in arsenic-mediated disease development and carcinogenesis during last ten years (2008–2018).

Results

The role of the specific histone marks in arsenic toxicity has been detailed. A comprehensive list that includes major arsenic-induced histone modifications identified for the last 10 years has been documented and details of different states of arsenic, organisms, exposure type, study platform, and findings were provided. An arsenic signature panel was suggested to help in early prognosis. An attempt has been made to identify the grey areas of research.

Prospects

Future prospective multi-target analyses of the inter-molecular crosstalk among different histone marks are needed to be explored further in order to understand the mechanism of arsenic toxicity and carcinogenicity and to confirm the suitability of these epi-marks as prognostic markers.

Introduction

Chromatin contains information that encodes the basic components to build-up an organism as a whole. With nearly 3.2 × 109 nucleotides making up the DNA content in the nucleus of each human cell alone (Brown, 2002), it is a massive factory of molecules that regulate required functions within the cell: metabolism, transport, cell division, repair, etc. Approximately 147 base pairs of DNA wrap around the histone octamer core composed of canonical histone proteins and their variants (Cosgrove and Wolberger, 2005). Approximately 20,000 genes are actively transcribed from the human genome, which accounts for 3300 mega base-pairs according to the recently updated database (GRCh38.p12, NCBI). Of the largest chromosomes, hChr-1 (human chromosome-1) has approximately 2000 protein-coding genes while the smallest hChr-Y has approximately 71 identified protein-coding genes; the hChr-19 is almost 1/4th time smaller than hChr-1, yet it has 1500 protein-coding genes. Thus, the size of a chromosome might not be positively correlated with the total number of genes that it can house and hence, it is possible that a highly orchestrated and stratified mechanism exists within this chromatin micro-environment for regulation of gene expression within the cell. In a very recent study, using single cell Hi-C, the authors showed how the chromatin compartmentalizes into an active and in-active segment, the former being closer to the nuclear membrane (Stevens et al., 2017). Accessibility to the chromatin to the interacting molecules (transcription factors, polymerases, ATP dependent chromatin remodeling complexes, etc.) ultimately determines the fate of the cell (Armstrong et al., 2018; Lamparter et al., 2017; Zhou et al., 2016). Further, epigenetic alteration of the histone and the DNA fine-tunes this accessibility. For example, N terminal histone tail plays a critical role in determining the epigenetic profile of a section of chromosome. Specific enzymatic activities chemically modify these tails like acetylation, sumoylation, ribosylation, ubiquitination, and methylation which may increase or decrease the activity of the genomic niche (Xu et al., 2017). To add on to these mechanisms, ATP dependent chromatin remodelers act like motors and identify the characteristic DNA and histone elements within the nucleosomes as well as extra-nucleosomal regions and translocate the nucleosomal core leading to alteration in accessibility of the chromatin (Paul and Bartholomew, 2018; Tyagi et al., 2016).

In this review, we refer to the array of histone post-translational modifications (PTMs) as the histone “code”, encrypting various functions of the cell-like transcriptional regulation, DNA damage repair and replication, progression of the cell cycle and its regulation and developmental processes processes (Cosgrove and Wolberger, 2005; Escargueil et al., 2008; Jenuwein and Allis, 2001; Paquin and Howlett, 2018; Williamson et al., 2012). Different cells have different ways to respond to certain cues within the micro-environment, leading to changes in histone signatures. An ensemble of these histone codes literally forms a “cypher” that are being regularly read by the chromatin-binding proteins that regulate normal as well as abnormal processes within the cells. For example, the change of a stem cell towards a more committed or differentiated form changes the global histone signatures significantly (Corley and Kroll, 2015). Owing to its covalent nature, these modifications are reversible and have been researched for targeted therapeutic strategies. For example, hypo-acetylation of H4 is associated with the breast cancer malignancy while acetylation of H3K9 has been associated with the hepatocellular carcinoma (Suzuki et al., 2009a). This dynamic property of the histone modifying enzymes has attracted much attention from the pharmaceutical companies in recent years (Liu et al., 2006; Oh et al., 2015; Rahman and Grundy, 2011). Unfortunately, not much is known about arsenic-induced alterations in theese post-translational modifications, although chronic exposure to arsenic affects nearly 150 million people in about 70 countries with ever increasing evidence of cancerous outcomes (Ahsan et al., 2006; Garcia-Esquinas et al., 2013; Mendez et al., 2017).

Arsenic, a classified group 1 carcinogenic metalloid, enters our system through environmental exposure (drinking water, dietary and/or occupational sources) leading to a plethora of human health disorders in the exposed population across the globe (WHO, 2010). A number of comprehensive reviews have described the mode of genotoxicity of arsenicosis: The major pathways identified are those involved in DNA damage repair pathway, cell cycle checkpoint regulators, oxidative stress mediators including nuclear and mitochondrial counterparts, etc. (Argos, 2015; Bhattacharjee et al., 2016, 2013a). In order to delineate the of role histone PTMs as a susceptibility factor and/or causal factor in arsenic toxicity, we have restricted our search on these major pathways: transcriptional regulation, oxidative stress, DNA damage repair pathway, and cell cycle check point regulators. Relevant search was conducted using keywords like “arsenic-induced histone post-translational modifications”, “transcriptional regulation”, “DNA damage”, “cell cycle check point regulators”, and “oxidative stress”. In this review, we will describe what is known about arsenic-induced toxicity but will focus mainly on the potential of identifying histone modifications as epigenetic biomarkers and on how epigenetic code alters with arsenic exposure.

Section snippets

Arsenic exposure: mobilization, accessibility and effects

Arsenic is abundant in various geographical hotspots. The exposure hotspots include countries or parts of countries such as Ganga Brahmaputra basin in Eastern India, Bangladesh, parts of China, Vietnam, Thailand, Argentina, and the United States of America. By definition, Group 1 carcinogen is such agent that has sufficient experimental evidence in animals and strong evidence in exposed humans that the agent acts through a relevant mechanism of carcinogenicity (IARC Monographs, 2019). Arsenic

Role of histone PTMs in arsenic-induced transcriptional regulation

The histone PTMs regulate the expression of a particular gene, which is cell-specific and can regulate the accessibility of the chromatin depending on the environmental cues. The transcription factors and RNA polymerases need an easy passage to anchor to the DNA backbone before transcribing the RNA. Certain histone PTMs have been associated with either actively transcribed gene promoters, enhancers, or inactive gene promoters. For example, H3K4me1/me3 are well characterized PTMs present at

Epigenetic signature as a potential diagnostic tool and future research prospects

Effective, accurate methods of early detection and clinical diagnosis are urgently required for risk assessment at the treatable stage since the hallmarks of arsenic toxicity develop after a long period of latency (more than 10 years) and 80-85% individuals in a population do not develop any phenotypic manifestation of these symptoms when exposed to arsenic (Ghosh et al., 2006). Emerging number of reports point toward the potential role of circulating cell-free nucleosomes (ccfn) and cell-free

Author contribution

P.B and Dr. P.B. conceived the idea, performed literature review and prepared the tables. P.B, S.P and Dr. P.B. wrote the manuscript. P.B and S.P prepared the figures.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Declaration of Competing Interest

The authors declare that they have no conflict of interest.

Acknowledgement

We thankfully acknowledge Dr. Kestas G. Bendinskas, Professor of Biochemistry, Department of Chemistry, SUNY- Oswego, for reviewing and carefully editing our manuscript.

References (118)

  • A.E. Escargueil et al.

    What histone code for DNA repair?

    Mutat. Res. - Rev. Mutat. Res.

    (2008)
  • Q. Feng et al.

    Methylation of H3-lysine 79 is mediated by a new family of HMTases without a SET domain

    Curr. Biol.

    (2002)
  • S.R. Gadhia et al.

    Trace metals alter DNA repair and histone modification pathways concurrently in mouse embryonic stem cells

    Toxicol. Lett.

    (2012)
  • Y. Ge et al.

    Inhibition of monomethylarsonous acid (MMAIII)-induced cell malignant transformation through restoring dysregulated histone acetylation

    Toxicology

    (2013)
  • Y. Ge et al.

    Mapping dynamic histone modification patterns during arsenic-induced malignant transformation of human bladder cells

    Toxicol. Appl. Pharmacol.

    (2018)
  • T. Gonzalez-Cortes et al.

    DNA methylation of extracellular matrix remodeling genes in children exposed to arsenic

    Toxicol. Appl. Pharmacol.

    (2017)
  • P. Halley et al.

    Regulation of the apolipoprotein gene cluster by a long noncoding RNA

    Cell Rep.

    (2014)
  • K.J. Herbert et al.

    Arsenic exposure disrupts epigenetic regulation of SIRT1 in human keratinocytes

    Toxicol. Appl. Pharmacol.

    (2014)
  • P. Hinhumpatch et al.

    Oxidative DNA damage and repair in children exposed to low levels of arsenic in utero and during early childhood: application of salivary and urinary biomarkers

    Toxicol. Appl. Pharmacol.

    (2013)
  • P. Hinhumpatch et al.

    Oxidative DNA damage and repair in children exposed to low levels of arsenic in utero and during early childhood: application of salivary and urinary biomarkers

    Toxicol. Appl. Pharmacol.

    (2013)
  • D.T. Huyen et al.

    Geological and geochemical characterizations of sediments in six borehole cores from the arsenic-contaminated aquifer of the Mekong Delta

    Vietnam. Data Brief.

    (2019)
  • M. Inagawa et al.

    Histone H3 lysine 9 methyltransferases, G9a and GLP are essential for cardiac morphogenesis

    Mech. Dev.

    (2013)
  • W.J. Jo et al.

    Acetylated H4K16 by MYST1 protects UROtsa cells from arsenic toxicity and is decreased following chronic arsenic exposure

    Toxicol. Appl. Pharmacol.

    (2009)
  • H.G. Kim et al.

    Polycomb (PcG) proteins, BMI1 and SUZ12, regulate arsenic-induced cell transformation

    J. Biol. Chem.

    (2012)
  • L. Li et al.

    Cross-talk between the H3K36me3 and H4K16ac histone epigenetic marks in DNA double-strand break repair

    J. Biol. Chem.

    (2017)
  • J. Liu et al.

    Dynamics of RNA polymerase II pausing and bivalent histone H3 methylation during neuronal differentiation in brain development

    Cell Rep.

    (2017)
  • T. Liu et al.

    Histone deacetylase inhibitors: multifunctional anticancer agents

    Cancer Treat. Rev.

    (2006)
  • L. Ma et al.

    Specific histone modification responds to arsenic-induced oxidative stress

    Toxicol. Appl. Pharmacol.

    (2016)
  • L. Martínez et al.

    Impact of early developmental arsenic exposure on promotor CpG-island methylation of genes involved in neuronal plasticity

    Neurochem. Int.

    (2011)
  • C. Mayr et al.

    The histone methyltransferase G9a: a new therapeutic target in biliary tract cancer

    Hum. Pathol.

    (2018)
  • Y. Niu et al.

    Oxidative stress alters global histone modification and DNA methylation

    Free Radic. Biol. Med.

    (2015)
  • S.Y. Oh et al.

    The histone methyltransferase inhibitor BIX01294 inhibits HIF-1α stability and angiogenesis

    Mol. Cells

    (2015)
  • S. Paul et al.

    Epimutagenesis: a prospective mechanism to remediate arsenic-induced toxicity

    Environ. Int.

    (2015)
  • R. Ruiz-Ramos et al.

    Sodium arsenite induces ROS generation, DNA oxidative damage, HO-1 and c-Myc proteins, NF-kappaB activation and cell proliferation in human breast cancer MCF-7 cells

    Mutat. Res.

    (2009)
  • V. Selvaraj et al.

    Arsenic trioxide (As(2)O(3)) induces apoptosis and necrosis mediated cell death through mitochondrial membrane potential damage and elevated production of reactive oxygen species in PLHC-1 fish cell line

    Chemosphere.

    (2013)
  • A. Stajnko et al.

    Arsenic metabolites; selenium; and AS3MT, MTHFR, AQP4, AQP9, SELENOP, INMT, and MT2A polymorphisms in Croatian-Slovenian population from PHIME-CROME study

    Environ. Res.

    (2019)
  • T. Suzuki et al.

    Trivalent dimethylarsenic compound induces histone H3 phosphorylation and abnormal localization of Aurora B kinase in HepG2 cells

    Toxicol. Appl. Pharmacol.

    (2009)
  • H. Ahsan et al.

    Arsenic exposure from drinking water and risk of premalignant skin lesions in Bangladesh: baseline results from the Health Effects of Arsenic Longitudinal Study

    Am. J. Epidemiol.

    (2006)
  • M. Argos

    Arsenic exposure and epigenetic alterations: recent findings based on the illumina 450K DNA methylation array

    Curr. Environ. Heal. reports.

    (2015)
  • R.L. Armstrong et al.

    Chromatin conformation and transcriptional activity are permissive regulators of DNA replication initiation in Drosophila

    Genome Res.

    (2018)
  • M. Banerjee et al.

    High arsenic in rice is associated with elevated genotoxic effects in humans

    Sci. Rep.

    (2013)
  • M. Bauden et al.

    Circulating nucleosomes as epigenetic biomarkers in pancreatic cancer

    Clin. Epigenet.

    (2015)
  • S.H. Bernhart et al.

    Changes of bivalent chromatin coincide with increased expression of developmental genes in cancer

    Sci. Rep.

    (2016)
  • A. Bhargava et al.

    Epigenetic biomarkers for risk assessment of particulate matter associated lung Cancer

    Curr. Drug Targets

    (2018)
  • A. Bhargava et al.

    Cell-free circulating epigenomic signatures: non-invasive biomarker for cardiovascular and other age-related chronic diseases

    Curr. Pharm. Des.

    (2017)
  • J. Brocato et al.

    Basic mechanics of DNA methylation and the unique landscape of the DNA methylome in metal-induced carcinogenesis

    Crit. Rev. Toxicol.

    (2013)
  • T.A. Brown

    Genomes

    (2002)
  • Y. Chervona et al.

    Associations between arsenic exposure and global posttranslational histone modifications among adults in Bangladesh

    Cancer Epidemiol. Biomarkers Prev.

    (2012)
  • S. Chitale et al.

    H4K20me2: orchestrating the recruitment of DNA repair factors in nucleotide excision repair

    Nucleus

    (2018)
  • I.A. Ciechomska et al.

    Pre-treatment or post-treatment of human glioma cells with BIX01294, the inhibitor of histone methyltransferase G9a, sensitizes cells to temozolomide

    Front. Pharmacol.

    (2018)
  • Cited by (11)

    • Prevention of inorganic arsenic induced squamous cell carcinoma of the skin in Swiss albino mice by black tea through epigenetic modulation

      2022, Heliyon
      Citation Excerpt :

      Arsenic mediates its carcinogenicity by excessive generation of reactive oxygen species (ROS) [2]. Metabolism of inorganic arsenic (iAs) utilises S-adenosyl methionine (SAM), whose depletion occurs due to chronic exposure to iAs, altering global methylation pattern of DNA and gene expression [4]. iAs is also known to induce different types of epigenetic modulations, and the present study focuses on post-translational histone modifications (PTM) including histone methylation, demethylation and acetylation.

    • Epigenetic modifications from arsenic exposure: A comprehensive review

      2022, Science of the Total Environment
      Citation Excerpt :

      Post-translational modifications (PTMS) of these histone proteins leading to acetylation, methylation, phosphorylation and others such as deamination, phosphoacetylation, nitrosylation, ADP ribosylation, proline isomerization, ubiquitylation and SUMoylation have been known to regulate the chromatin biology and gene expression and repression (Roy et al., 2015; Xu et al., 2017). Histone modifications not only regulate chromatin structure and play an important role in transcriptional control but also affect other processes like replication, repair and recombination (Bhattacharjee et al., 2020). Arsenic is one of the metalloids which is able to induce carcinogenesis by bringing about chromosomal instability via induction of structural perturbations in higher order chromatin structures (Martinez-Zamudio and Ha, 2011).

    • Depletion of S-adenosylmethionine pool and promoter hypermethylation of Arsenite methyltransferase in arsenic-induced skin lesion individuals: A case-control study from West Bengal, India

      2021, Environmental Research
      Citation Excerpt :

      Higher utilization and depletion of SAM pool is associated with hypermethylation and down-regulation of tumour suppressor genes (Lozano-Rosas et al., 2020; Lu et al., 2020; Wang et al., 2017). Epimutagenic effects of arsenic result in altered gene expression and downstream protein functionality through epigenetic modulation involving SAM utilization (Bhattacharjee et al., 2019; Bhattacharjee and Paul, 2019; Das et al., 2019; Paul et al., 2015; Zhou et al., 2018; Kietzmann et al., 2017). Arsenic-induced modifications in DNA methylation, post-translational histone modifications and altered expression profile of micro-RNAs leading to increased carcinogenicity is well-established (Bhattacharjee et al., 2019; Winterbottom et al., 2019; Cheng et al., 2018; Sanyal et al., 2020a, 2020b; Engström et al., 2013).

    View all citing articles on Scopus
    View full text