Nrf2 protects against As(III)-induced damage in mouse liver and bladder

https://doi.org/10.1016/j.taap.2009.06.010Get rights and content

Abstract

Arsenic compounds are classified as toxicants and human carcinogens. Environmental exposure to arsenic imposes a big health issue worldwide. Arsenic elicits its toxic efforts through many mechanisms, including generation of reactive oxygen species (ROS). Nrf2 is the primary transcription factor that controls expression of a main cellular antioxidant response, which is required for neutralizing ROS and thus defending cells from exogenous insults. Previously, we demonstrated a protective role of Nrf2 against arsenic-induced toxicity using a cell culture model. In this report, we present evidence that Nrf2 protects against liver and bladder injury in response to six weeks of arsenic exposure in a mouse model. Nrf2−/− mice displayed more severe pathological changes in the liver and bladder, compared to Nrf2+/+ mice. Furthermore, Nrf2−/− mice were more sensitive to arsenic-induced DNA hypomethylation, oxidative DNA damage, and apoptotic cell death. These results indicate a protective role of Nrf2 against arsenic toxicity in vivo. Hence, this work demonstrates the feasibility of using dietary compounds that target activation of the Nrf2 signaling pathway to alleviate arsenic-induced damage.

Introduction

Groundwater contaminated with arsenic is the major source of human exposure to arsenic. Chronic arsenic poisoning imposes a global health issue (Tchounwou et al., 2003, Walvekar et al., 2007). Therefore, the World Health Organization (WHO) has set the standard limit for arsenic to 10 parts per billion (ppb) (Kayajanian, 2003). However, approximately 57 million people are drinking groundwater with arsenic concentrations above 10 ppb. Arsenic has caused poisonings in Bangladesh, Bengal, Thailand, Finland, Hungary, Chile, Taiwan, Vietnam, Cambodia, Mexico, Argentina, and China, where geological environments are conducive to generate high amounts of arsenic compounds in groundwater (Smith et al., 2000, Kayajanian, 2003, Tchounwou et al., 2003). Furthermore, many states within the United States also have significant concentrations (up to 50 ppm) of arsenic in the groundwater (Tchounwou et al., 2003, Knobeloch et al., 2006).

The most common species of arsenic in groundwater are arsenate (As(V)) and arsenite (As(III)). Arsenic is able to undergo redox conversion between As(V) and As(III). Following uptake from drinking water, inorganic arsenic species are converted in the liver into methylated arsenic species that are excreted from the bladder. Thus, the bladder is the major target organ that is exposed to methylated arsenic species in populations who consume arsenic-contaminated water (Tchounwou et al., 2003, Kenyon et al., 2005). Chronic exposure to arsenic can cause skin, lung and bladder cancers (Hsueh et al., 1995, Brown et al., 1997, Cohen et al., 2000, Smith et al., 2006). A small but measurable increase in the incidence of bladder cancer was associated with exposure to concentration as low as 10 ppm of inorganic arsenic (Chu and Crawford-Brown, 2006). In addition to cancers, epidemiological studies have also established a strong correlation between chronic arsenic exposure and various non-cancer human diseases, such as hyperkeratosis, atherosclerosis, diabetes, and chronic obstructive pulmonary diseases (Tseng, 2002, Tchounwou et al., 2003, Navas-Acien et al., 2006).

The transcription factor, Nrf2, controls a cellular antioxidant response through transcriptional upregulation of an array of downstream genes, such as glutamate cysteine ligase (GCL), heme oxygenase-1 (HO-1), glutathione s-trasferase (GST), multidrug-resistance proteins (MRPs), and NAD(P)H quinone oxidoreductase-1 (NQO1) (Lau et al., 2008, Hayes and McMahon, 2009). Coordinated upregulation of the Nrf2-mediated endogenous antioxidants, phase II detoxifying enzymes, and drug transporters is essential in defending cells or animals from damage by environmental insults. This has been clearly demonstrated in many studies using Nrf2−/− mice. Nrf2−/− mice displayed a greater sensitivity to toxic effects induced by benzo[a]pyrene, diesel exhaust, pentachlorophenol, and bleomycin (Aoki et al., 2001, Ramos-Gomez et al., 2001, Cho et al., 2004, Umemura et al., 2006).

Arsenic exerts its toxicity in part by generation of ROS (Hei et al., 1998, Kitchin and Ahmad, 2003, Liu et al., 2003, Das et al., 2005). Consistent with the role of ROS in arsenic toxicity/carcinogenicity, endogenous sulfhydryl groups and the non-protein sulfhydryl glutathione (GSH) have been reported to play an important role in detoxification of arsenic (Duyndam et al., 2001). The exogenous antioxidant N-acetylcysteine is also able to prevent arsenic-induced toxicity (Liu et al., 2003). Arsenic itself was reported to induce the Nrf2-dependent antioxidant response, although the detailed mechanism of Nrf2 induction by arsenic remains to be explored (He et al., 2006, Wang et al., 2008). Numerous studies have been performed to elucidate events associated with arsenic-induced damage, both in animal and cell culture models. Results from these studies have revealed that arsenic induces multiple biological effects, such as DNA damage, apoptotic cell death, and global DNA hypomethylation, which represent both genotoxic and non-genotoxic mechanisms of action (Huang et al., 1999, Kirkpatrick et al., 2003, Chen et al., 2004). Methylation is a covalent modification of genomic DNA and occurs on cytosines followed by guanines (CpG). DNA methylation appears to influence gene expression through alteration in chromatin structure and in DNA protein interaction. An overall decrease in the content of 5-methylcytosine (5-mc) in DNA has been viewed as an early event in tumor formation in humans and animal models (Lapeyre et al., 1981, Jones and Buckley, 1990, Umemura et al., 1990).

Recently, by using both genetic and biochemical approaches, we demonstrated a protective role of Nrf2 against arsenic-induced toxicity in a cell culture model (Wang et al., 2007). In vivo, even though Nrf2−/− mice have been demonstrated to display increased sensitivity to chemical toxicants and carcinogens (Chan and Kan, 1999, Aoki et al., 2001, Enomoto et al., 2001, Ramos-Gomez et al., 2001, Cho et al., 2002, Yu and Kensler, 2005), currently no study has been carried out to determine the effect of Nrf2 deficiency on arsenic-induced toxicity and carcinogenicity. In this report, we hypothesized that Nrf2−/− mice are more vulnerable to the arsenic-induced toxicity, compared to Nrf2+/+ mice. Our results confirmed that Nrf2−/− mice were more sensitive to arsenic-induced DNA hypomethylation, oxidative DNA damage, and apoptotic cell death. Furthermore, more severe pathological alterations were observed in the liver and bladder of Nrf2−/− mice, compared to Nrf2+/+ mice.

Section snippets

Animals and treatments

Nrf2−/− mice were originally generated in Dr. Kan's laboratory (University of California, San Francisco). Mice were housed in polycarbonate cages (4/cage), provided AIN-76A diet and water ad libitum and maintained on a 12–12 h light–dark cycle at 22 ± 5°C and 50 ± 20% relative humidity. At 8 weeks of age, Nrf2+/+ and Nrf2−/− mice (4 mice per group) were treated with 1 ppm, 10 ppm or 100 ppm of sodium arsenite (As(III), Sigma, St Louis, MO, USA) through drinking water for six weeks. All 32 mice

There was no difference in body weight or water consumption between Nrf2+/+ and Nrf2−/− mice

All 32 mice survived the 6-week As(III) treatment, with no obvious systemic toxicity, such as diarrhea, hair loss, or nasal hemorrhaging. Body weight gained during the 6-week period was recorded at the day of harvest. The weight gained was reduced as the doses of As(III) increased (Fig. 1A). In addition, no difference in weight gained between Nrf2+/+and Nrf2−/− mice was observed (Fig. 1A). The amount of water intake in Nrf2+/+ and Nrf2−/− mice was similar, except that the Nrf2+/+ and Nrf2−/−

Discussion

Many previous findings suggest that an adaptive response exists in mammals, i.e. previous exposure to low toxic reagents can elicit a beneficial effect that reduces damage by subsequent insults. We postulated that the adaptive response is derived from activation of the Nrf2 pathway based on the following observations: (i) resveratrol and epigallocatechin gallate, two known inducers of Nrf2, prevented arsenite-induced transformation of human osteogenic sarcoma cells. (Yang et al., 2005). (ii)

Acknowledgments

This study was supported by the NIH grant ES015010 and ACS grant #RSG-07-154-01-CNE to D. D. Zhang, and an NIH center grant ES 006694.

References (47)

  • LiuL. et al.

    Oxidative stress contributes to arsenic-induced telomere attrition, chromosome instability, and apoptosis

    J. Biol. Chem.

    (2003)
  • PottW.A. et al.

    Antagonistic interactions of an arsenic-containing mixture in a multiple organ carcinogenicity bioassay

    Cancer Lett.

    (1998)
  • SnowE.T. et al.

    Arsenic, mode of action at biologically plausible low doses: what are the implications for low dose cancer risk?

    Toxicol. Appl. Pharmacol.

    (2005)
  • WangX.J. et al.

    Nrf2 protects human bladder urothelial cells from arsenite and monomethylarsonous acid toxicity

    Toxicol. Appl. Pharmacol.

    (2007)
  • WangX.J. et al.

    Activation of Nrf2 by arsenite and monomethylarsonous acid is independent of Keap1–C151: enhanced Keap1–Cul3 interaction

    Toxicol. Appl. Pharmacol.

    (2008)
  • YangC. et al.

    Caffeic acid phenethyl ester (CAPE) prevents transformation of human cells by arsenite (As) and suppresses growth of As-transformed cells

    Toxicology

    (2005)
  • YuX. et al.

    Nrf2 as a target for cancer chemoprevention

    Mutat. Res.

    (2005)
  • BaeD.S. et al.

    Characterization of gene expression changes associated with MNNG, arsenic, or metal mixture treatment in human keratinocytes: application of cDNA microarray technology

    Environ. Health Perspect.

    (2002)
  • BrownK.G. et al.

    Skin cancer and inorganic arsenic: uncertainty-status of risk

    Risk Anal.

    (1997)
  • ChanK. et al.

    Nrf2 is essential for protection against acute pulmonary injury in mice

    Proc. Natl. Acad. Sci. U. S. A.

    (1999)
  • ChenH. et al.

    Chronic inorganic arsenic exposure induces hepatic global and individual gene hypomethylation: implications for arsenic hepatocarcinogenesis

    Carcinogenesis

    (2004)
  • ChoH.Y. et al.

    Role of NRF2 in protection against hyperoxic lung injury in mice

    Am. J. Respir. Cell Mol. Biol.

    (2002)
  • ChoH.Y. et al.

    The transcription factor NRF2 protects against pulmonary fibrosis

    FASEB J.

    (2004)
  • Cited by (78)

    • Mechanism for arsenic-induced toxic effects

      2023, Handbook of Arsenic Toxicology
    • Protective actions of nuclear factor erythroid 2-related factor 2 (NRF2) and downstream pathways against environmental stressors

      2022, Free Radical Biology and Medicine
      Citation Excerpt :

      Also, they demonstrated that this upregulation is independent of the previously identified cysteine residue C151 KEAP1, which indicates a specific mechanism by arsenic compared to other NRF2 inducers [268]. On the other hand, it has been demonstrated that autophagy could be attenuated by SFN and tert-butylhydroquinone (tBHQ) supporting that the KEAP1-C151-dependent mechanism of NRF2 activation is protective, and they could be a therapy against arsenic toxicity [270,272,276,277]. Cadmium (Cd) is an especially toxic and persistent heavy metal and a known carcinogen [278,279].

    View all citing articles on Scopus
    1

    These authors contribute equally to this work.

    View full text