Abstract
Chronic human exposure to inorganic arsenic (iAs), a potent environmental oxidative stressor, is associated with increased prevalence of Type 2 diabetes, where impairment of pancreatic β-cell function is a key pathogenic factor. Nuclear factor E2-related factor 2 (Nrf2) is a central transcription factor regulating cellular adaptive response to oxidative stress. However, persistent activation of Nrf2 in response to chronic oxidative stress, including inorganic arsenite (iAs3+) exposure, blunts glucose-triggered reactive oxygen species (ROS) signaling and impairs glucose-stimulated insulin secretion (GSIS). In the current study, we found that MIN6 pancreatic β-cells with stable knockdown of Nrf2 (Nrf2-KD) by lentiviral shRNA and pancreatic islets isolated from Nrf2-knockout (Nrf2−/−) mice exhibited reduced expression of several antioxidant and detoxification enzymes in response to acute iAs3+ exposure. As a result, Nrf2-KD MIN6 cells and Nrf2−/− islets were more susceptible to iAs3+ and monomethylarsonous acid (MMA3+)-induced cell damage, as measured by decreased cell viability, augmented apoptosis and morphological change. Pretreatment of MIN6 cells with Nrf2 activator tert-butylhydroquinone protected the cells from iAs3+-induced cell damage in an Nrf2-dependent fashion. In contrast, antioxidant N-acetyl cysteine protected Nrf2-KD MIN6 cells against acute cytotoxicity of iAs3+. The present study demonstrates that Nrf2-mediated antioxidant response is critical in the pancreatic β-cell defense mechanism against acute cytotoxicity by arsenic. The findings here, combined with our previous results on the inhibitory effect of antioxidants on ROS signaling and GSIS, suggest that Nrf2 plays paradoxical roles in pancreatic β-cell dysfunction induced by environmental arsenic exposure.
Keywords: arsenic, diabetes, pancreatic β-cell, islets, Nrf2, oxidative stress, cytotoxicity
Introduction
There is growing evidence that chronic exposure to inorganic arsenic (iAs) is associated with increased prevalence of Type 2 diabetes (T2D), a metabolic disease that is attributed to a combination of genetic, lifestyle and environmental factors (Lai et al., 1994; Rahman et al., 1998; Wang et al., 2003; Tseng, 2004; Nabi et al., 2005; Chiu et al., 2006; Coronado-Gonzalez et al., 2007; Meliker et al., 2007; Navas-Acien et al., 2008; Navas-Acien et al., 2009; Del Razo et al., 2011). However, the mode of action for the diabetogenic effect of iAs is largely unknown. The development of T2D is underpinned by a progressive worsening of insulin responsiveness and pancreatic β-cell function (Lowell and Shulman, 2005; Robertson and Harmon, 2006). Normal pancreatic β-cells can compensate for attenuated insulin sensitivity by increasing insulin secretion and/or β-cell mass (Prentki and Nolan, 2006). Insufficient compensation ultimately leads to the onset of glucose intolerance and T2D (Kajimoto and Kaneto, 2004). Amidst the various mechanisms proposed for β-cell dysfunction and their roles in the progression to diabetes, oxidative stress is a common denominator (Evans et al., 2003; Kajimoto and Kaneto, 2004; Robertson and Harmon, 2006).
Nuclear factor E2-related factor 2 (Nrf2, also known as Nfe2l2) is a master regulator of the cellular adaptive response to oxidative and electrophilic stress (Maher and Yamamoto, 2010; Pi et al., 2010b). In response to oxidative/electrophilic stress, Nrf2 heterodimerizes with small Maf proteins and other basic leucine zipper proteins, binding to antioxidant response elements (AREs) in the promoters of many phase II detoxification and antioxidant genes to increase their transcription (Maher and Yamamoto, 2010). Evidence supporting the pivotal roles of Nrf2 in protecting against oxidative/electrophilic stress comes, in part, from studies conducted in Nrf2-knockout (Nrf2−/−) mice and Nrf2-knockdown (Nrf2-KD) cells. These animals and cells exhibit a severe deficiency in the coordinated gene regulatory program for adaptive antioxidant response, resulting in a high susceptibility to oxidative stress-related disorders, chemical carcinogenesis and cytotoxicity (Itoh et al., 2010; Liu et al., 2010; Zhao et al., 2012). Thus, the Nrf2-mediated antioxidant response represents a critically important cellular defense mechanism that serves to maintain intracellular redox homeostasis and limit oxidative damage. iAs and its methylated trivalent metabolites, including monomethylarsonous acid (MMA3+) and dimethylarsinous acid (DMA3+), are potent oxidative stressors (Pi et al., 2002; Pi et al., 2003a; Pi et al., 2003b; Bailey et al., 2010). Acute exposure to inorganic arsenite (iAs3+) and MMA3+ has been demonstrated to activate Nrf2-mediated adaptive antioxidant response in a variety of human and rodent cells (Pi et al., 2003b; Pi et al., 2007a; Pi et al., 2008; Wang et al., 2008; Zhao et al., 2011; Zhao et al., 2012). Our previous studies (Pi et al., 2007b; Fu et al., 2011) reported that up to 96 hrs of exposure of pancreatic β-cell line INS-1(832/13) cells and isolated mouse islets to low levels of iAs3+ led to decreased glucose-stimulated insulin secretion (GSIS), which is associated with adaptive induction of antioxidant enzymes, including catalase and glutathione peroxidase. However, the precise role of Nrf2 in arsenic-induced β-cell dysfunction is still unclear. Here we report that Nrf2-mediated antioxidant response plays a critical role in the pancreatic β-cell defense mechanism against the acute cytotoxicity induced by high-concentrations of iAs3+ and MMA3+. Given the important role of reactive oxygen species (ROS) signaling in GSIS (Pi et al., 2007b), the present study and our previous findings on the inhibitory effect of antioxidants on ROS signaling (Pi et al., 2007b; Fu et al., 2011) suggest that Nrf2 plays paradoxical roles in pancreatic β-cell dysfunction induced by environmental arsenic exposure.
Materials and Methods
Cell culture and reagents
MIN6 cells were kindly provided by Dr. Marcia Haigis (Harvard University, Boston, MA) and maintained in Dulbecco’s modified Eagle’s medium (DMEM) containing 25 mM glucose, with 15% fetal bovine serum (FBS), 100 units/ml penicillin, 100 µg/ml streptomycin, 2 mM L-glutamine and 5 µl/L β-mercaptoethanol in humidified 5% CO2, 95% air at 37°C. Culture media, FBS, phosphate buffered saline (PBS, pH7.4) and supplements were purchased from Invitrogen (Carlsbad, CA). Sodium arsenite, β-mercaptoethanol, tert-Butylhydroquinone (tBHQ), sulforaphane (SFN) and N-acetyl cysteine (NAC) were obtained from Sigma (St. Louis, MO). Methylarsine oxide (CH3AsO) was synthesized by Dr. William R. Cullen (University of British Columbia, Vancouver, Canada) using a method described previously (Cullen et al., 1989; Petrick et al., 2001).
Lentiviral-based shRNA transduction
MISSION shRNA lentiviral vectors were obtained from Sigma and lentiviral particles were prepared using the manufacturer's protocol. Lentiviral transduction of MIN6 cells with particles for shRNAs targeting Nrf2 (SHVRSNM_010902) or Scrambled (Scr) non-target negative control (SHC002V) was performed as described previously (Pi et al., 2010a). Briefly, 24 hrs prior to transduction, MIN6 cells were plated in 6-well plates at 40–50% confluency in complete medium described above. The following day, hexadimethrine bromide (Sigma), a transduction enhancer, was added to each well at a concentration of 8 µg/ml and viral particles were added to each well at a concentration of 2×105 transducing units (TU) per ml. Following a 24-hr incubation period, medium containing viral particles was removed and replaced with fresh medium containing 1 µg/ml of puromycin. Cells were grown to 90% confluence and sub-cultured in medium containing puromycin (1 µg/m).
Animals
Nrf2−/− mice developed as described previously (Itoh et al., 1997) were kindly provided by Dr. Masayuki Yamamoto (Tohoku University, Japan). The mice were backcrossed onto the C57BL/6J background for 7 generations using alternating male and female stock mice from The Jackson Laboratories (Bar Harbor, ME). The resulting wild-type (Nrf2+/+) and Nrf2−/− mice were used in the present study. The mice were housed in virus-free facilities on a 12-hr light/12-hr dark cycle and were fed NIH07 chow diet (Zeigier Brothers Gardners, PA) and provided with reverse osmosis water ad libitum. Genotyping was performed by PCR (primer sequences in Online Material Table S1) using genomic DNA that was isolated from tail snips. All protocols for animal use were approved by the Institutional Animal Care and Use Committee of The Hamner Institutes and were in accordance with the National Institutes of Health guidelines.
Islet isolation and primary culture
Pancreatic islets were isolated from 9–12 week-old mice by collagenase P (Roche, Switzerland) digestion, as described previously (Pi et al., 2007b). Islets were cultured in RPMI 1640 supplemented with 10 mM glucose, 10% FBS, 25 mM HEPES, 2 mM L-glutamine, 100 U/ml penicillin and 100 µg/ml streptomycin. The islets were cultured for 48 hrs before arsenic treatment.
Reverse transcription quantitative real-time PCR (RT-qPCR)
Total RNA in MIN6 cells was isolated with TRIzol reagent (Invitrogen) and subsequently subjected to cleanup using an RNase-Free DNase Set and RNeasy Mini Kit (Qiagen, Valencia, CA). Total RNA in cultured islets (500 islets each condition) was isolated using the RNeasy Mini Kit. Total RNA was reverse transcribed with MuLV reverse transcriptase and Oligo d(T) primers (Applied Biosystems, Foster City, CA). A SensiFAST SYBR Hi-ROX kit (BIOLINE USA Inc, Taunton, MA) was used for qPCR. The primers were designed using Primer Express 4 (Applied Biosystems) and synthesized by Bioneer Inc. (Alameda, CA). The primer sequences are listed in Table S3. Relative differences in gene expression among groups were determined from quantification cycle (Cq) values. These values were first normalized to 18 ribosomal RNA (18S) in the same sample (ΔCq) and expressed as the fold-change over control (2−ΔΔCq). Real-time fluorescence detection was performed using an ABI PRISM 7900HT Fast Real-time PCR System (Applied Biosystems). Details on the procedures of RNA quantification and RT-qPCR are described in Online Supplemental Materials.
Western blot analysis
Isolation of cell fractions and immunoblotting were performed as detailed previously (Pi et al., 2003b; Woods et al., 2009). An antibody for Nrf2 (sc-13032; 1:500) was obtained from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA). Antibodies for caspase-3 (#9662, 1: 1000) and cleaved caspase 3 (#9661, 1:1000) were purchased from Cell Signaling Technology, Inc. (Danvers, MA). Antibodies against β-ACTIN (A1978; 1:2000) and LAMIN A (L1293; 1:2500) were from Sigma.
Measurement of cell viability and caspase-3/7 activity
Cell viability was determined by using Non-Radioactive Cell-Proliferation Assay Kit (Promega, Madison, WI) as detailed previously (Pi et al., 2005). Caspase-3/7 activity was measured using the ApoLive-Glo Multiplex Assay Kit (Promega, Madison, WI) per the manufacturer's recommendation. A minimum of 5 replicates of 10,000 cells per well were plated in 96-well plates and allowed to adhere to the plate for 24 hrs, after which the media was removed and replaced with fresh media containing various concentrations of arsenicals. Cells were subsequently incubated for an additional 24 hrs, and cell viability and caspase-3/7 activity were determined.
Cell death assessment by flow cytometry
MIN6 cells were seeded in a 6-well plate and grown to approximately 80% confluence. After the cells were treated with iAs3+ for 24 hrs, the floating and attached cells were harvested for apoptosis analysis. Apoptotic and necrotic cells were analyzed by flow cytometry (FACS CantoII with HTS option, BD Biosciences) using the TACS Annexin V-FITC Apoptosis Detection Kit (Trevigen, Gaithersburg, MD) as detailed previously (Pi et al., 2005). For each sample, 10,000 cells were examined. The percentage of apoptotic and necrotic cells was determined by statistical analysis of the various dot plots by using CellQuest software (Diva 6.0, BD Biosciences).
Terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) staining
TUNEL staining of islets was performed using an ApopTag Plus Fluorescein In Situ Apoptosis Detection Kit (EMD Millipore, Billerica, MA). Briefly, following a 48-hr culture period, isolated islets were treated with iAs3+ for 24 hrs. Subsequently, the islets were washed with PBS and fixed for 24 hrs at room temperature in 2% (v/v) formaldehyde. After washing in PBS, islets were embedded in paraffin, sectioned, de-paraffinized and re-hydrated as described previously (Pi et al., 2009). The slides were then treated with proteinase K (20 µg/ml) for 15 min and incubated in 3% (v/v) H2O2 for 5 min to quench endogenous peroxidase activity. The slides were labeled by incubating for 60 min at 37 °C in the presence of terminal deoxynucleotidyl transferase (TdT) and for another 30 min in an anti-digoxigenin antibody conjugated with horseradish peroxidase. Negative controls were also prepared in which TdT treatment was omitted. Following TUNEL staining, slides were mounted with the Prolong Gold antifade reagent with DAPI (P36931, Invitrogen) and covered with coverslips and examined using an Axio Observer Z1 fluorescence microscope (Carl Zeiss, Inc. Oberkochen, Germany).
Statistical analyses
All statistical analyses were performed by using Graphpad Prism 4 (GraphPad Software, San Diego, CA), with p < 0.05 taken as significant. More specific indices of statistical significance are indicated in individual figure legends. Data are expressed as mean ± SD. For comparisons between two groups, a Student’s t-test was performed. For comparisons among three or more groups, one-way or two-way ANOVA with Bonferroni post hoc testing was performed.
Results
Stable knockdown of Nrf2 in MIN6 cells reduces the adaptive antioxidant response induced by iAs3+
In MIN6 cells, transduction of lentiviral shRNA against mouse Nrf2 (termed Nrf2-KD) significantly attenuated the mRNA expression of Nrf2 compared to Scr control cells (Fig. 1A). The effectiveness of Nrf2 silencing was confirmed by notably diminished protein expression of Nrf2 in cells challenged with known Nrf2 activators, including iAs3+, tBHQ and SFN (Fig. 1B). To determine the acute effect of iAs3+ exposure on Nrf2-mediated antioxidant response in pancreatic β-cells, the time-course (Fig.1C and D) and concentration-response (Fig. 1E and F) of Nrf2 expression in response to iAs3+ exposure were determined at mRNA (Fig.1C and E) and protein (Fig.1D and F) levels in Scr and Nrf2-KD cells. In contrast to a late induction of mRNA expression of Nrf2 (Fig. 1C), the protein levels of Nrf2 were elevated quickly and peaked between 2–6 hrs in Scr cells (Fig. 1D). Importantly, in response to a 6-hr exposure to iAs3+, the protein expression of Nrf2 in Scr cells was increased in a concentration-dependent manner. In contrast, Nrf2-KD cells expressed remarkably reduced levels of Nrf2 protein under basal and iAs3+-exposed conditions (Fig. 1F). In addition, acute iAs3+ exposure led to increased protein accumulation of Nrf2 in the nuclear fractions of Scr cells in a concentration-dependent fashion. Nrf2-KD cells showed a substantial reduction in iAs3+-induced Nrf2 nuclear accumulation (Fig. 2A). Furthermore, the expression of ARE-dependent genes, including NAD(P)H quinone oxidoreductase 1 (Nqo1), heme oxygenase 1 (Hmox-1), glutamate-cysteine ligase catalytic (Gclc) and sulfiredoxin (Srx) was substantially attenuated by Nrf2 silencing, indicating that Nrf2-mediated transcription was suppressed in Nrf2-KD cells (Fig. 2B and C).
Fig. 1.
Stable knockdown of Nrf2 results in reduced expression of Nrf2 in MIN6 cells. (A) mRNA expression of Nrf2 in MIN6 cells transduced with shRNA lentivirus targeted against mouse Nrf2 or Scrambled non-target negative control (Scr). (B) Reduced protein expression of Nrf2 in Nrf2-KD cells under basal and iAs3+ (5 µM), tert-Butylhydroquinone (tBHQ, 50 µM) or sulforaphane (SFN, 10 µM)-treated conditions. Cells were treated with the chemicals for 6 hrs. Whole cell lysates were used for immunoblotting, and β-ACTIN was used as a loading control. Vehicle, medium. (C and D) Time course of iAs3+-induced Nrf2 mRNA (C) and protein (D) expression. Cells were exposed to 4 µM iAs3+ for the indicated time. (E and F) Concentration-response of iAs3+-induced Nrf2 mRNA (E) and protein (F) expression after cells were exposed to iAs3+ for 6 hrs. Values in A, C and E are mean ± SD. n = 3–6. *p < 0.05 vs. Scr with the same treatment.
Fig. 2.
Nrf2 silencing results in reduced antioxidant response induced by iAs3+ in MIN6 cells. (A) Nrf2 protein levels in nuclear fractions of MIN6 cells. Lamin A was used as a loading control. (B and C) Lack of Nrf2 significantly reduces the expression of ARE-dependent genes. The concentration of iAs3+ used in B is 5 µM. The time point at which the measurements were made in C is 6 hrs. The expression of Nqo1, Hmox-1, Gclc and Srx was measured by real-time RT-PCR. The number in brackets following the gene name is the Cq value of that gene in Scr cells with vehicle. Values in B and C are means ± SD. n = 3. *p < 0.05 vs. Scr with the same treatment.
Silencing of Nrf2 sensitizes MIN6 cells to iAs3+ and CH3AsO-induced acute cytotoxicity
To study the protective effect of Nrf2 against the cytotoxicity of arsenic in pancreatic β-cells, we investigated the effect of stable knockdown of Nrf2 on the susceptibility to arsenic-induced cytotoxicity in MIN6 cells. As expected, Nrf2 silencing rendered the cells more susceptible to iAs3+-induced reduction in cell viability (Fig. 3A). This sensitization to iAs3+-induced cytotoxicity was further confirmed by measurement of apoptosis and necrosis using flow cytometry with Annexin V-FITC and propidium iodide (PI) double staining (Fig. 3C and D), as well as the expression and activity of Caspase 3 and/or 7 (Fig. 3E and F). It is generally accepted that iAs, including iAs3+ and iAs5+, are metabolized in humans by enzymatic and non-enzymatic mechanisms (Aposhian, 1997; Hayakawa et al., 2005) into monomethylarsonous acid (MMA3+), momomethylarsonic acid (MMA5+), dimethylarsinous acid (DMA3+) and dimethylarsinic acid (DMA5+). MMA (MMA3+ + MMA5+) and DMA (DMA3+ + DMA5+) are the major metabolites of iAs in human blood and urine (Pi et al., 2002). In addition, high levels of MMA and DMA were detected in the pancreas of mice with chronic iAs exposure (Paul et al., 2011), suggesting that methylated metabolites of iAs may be involved in environmental iAs exposure-induced β-cell dysfunction. Thus, the cytotoxic effect of MMA3+ was determined in MIN6 cells. As shown in Fig. 3B, CH3AsO exhibited much more of a potent inhibitory effect on cell viability than iAs3+ in MIN6 cells. In particular, Nrf2-KD cells exhibited significantly higher sensitivity than Scr to CH3AsO-induced cytotoxicity.
Fig. 3.
Stable knockdown of Nrf2 increases the susceptibility of MIN6 cells to iAs3+- and MMA3+-induced cytotoxicity. (A and B) Nrf2-KD cells are more sensitive to the cytotoxicity of iAs3+ (A) and MMA3+ (B). Cell viability was assessed by ApoLive-Glo Multiplex Assay following a 24-hr treatment with the indicated concentrations of arsenicals. (C) Representative plots of apoptosis measurements by flow cytometry. MIN6 cells stained with Annexin V and PI before being analyzed. FL, fluorescence. (D) Quantification of iAs3+-induced apoptosis by flow cytometry. Cells were exposed to iAs3+ for 24 hrs. Annexin V-positive cells were quantified as apoptotic cells. (E) Immunoblotting of Caspase-3 and cleaved-Caspase-3. Cells were exposed to iAs3+ for 24 hrs. Whole cell lysates were used for analysis, and β-ACTIN was used as a loading control. Vehicle, medium. (F) The activity of Caspase 3/7 measured by the Apolive-Glo Multiplex Assay in MIN6 cells exposed to iAs3+ for 24 hrs. Values in A, B, D and F are means ± SD. n = 3–6. *p < 0.05 vs. Scr with the same treatment.
Nrf2−/− islets show abrogated antioxidant response and are more sensitive to iAs3+-induced acute cytotoxicity
To validate the major findings in MIN6 cells, the Nrf2-mediated antioxidant response and cytotoxicity induced by acute iAs3+ exposure were determined in isolated mouse islets. As shown in Fig. 4A, acute iAs3+ exposure increased the expression of Nrf2 and its downstream genes, including Nqo1, Hmox-1, Gclc and Srx, in Nrf2+/+ islets. Deficiency of Nrf2 resulted in significantly reduced expression of most of these ARE-dependent genes. Following a 24-hr iAs3+ (20 or 50 µM) exposure, many islets lost their intact membranes and dispersed into disassociated cells and/or cell debris (Fig. 4B and C), indicating the cytotoxicity of iAs3+. Compared with Nrf2+/+, Nrf2−/− islets exhibited substantially reduced amounts of intact islets left after iAs3+ exposure (Fig. 4D). In addition, TUNEL staining demonstrated that Nrf2−/− islets were more sensitive to iAs3+-induced apoptosis (Fig. 5).
Fig. 4.
Deficiency of Nrf2 results in reduced antioxidant response and increased sensitivity to acute iAs3+ exposure-induced cytotoxicity in primary cultured mouse islets. (A) Nrf2−/− islets exhibit reduced expression of ARE-dependent genes. Isolated islets were exposed to 5 µM iAs3+ for 6 hrs followed by immediate isolation of total RNA and real-time RT-PCR measurement for Nqo1, Hmox-1, Gclc and Srx. The number in brackets following the gene name is the Cq value of that gene in Nrf2+/+ islets with Vehicle. n = 3. *p < 0.05 vs. the same genotype with vehicle;#p < 0.05 vs. Nrf2+/+ islets with the same treatment. (B) Representative images of cultured islets following a 24-hr treatment with indicated concentrations of iAs3+. n = 3. The islets were examined by the Olympus SZX7 Zoom Stereomicroscope (4 ×). (C) Representative images of cultured islets following a 24-hr iAs3+ (10 µM) treatment. The islets were examined by the Carl Zeiss Axiovert 40 CFL inverted microscope (10 ×). (D) Quantification of intact islets that remained following a 24-hr iAs3+ exposure as in (B).
Fig. 5.
(A) Representative TUNEL staining in cultured islets which were incubated for 24 h in the presence or absence of 10 µM of iAs3+. red = DAPI; green = TUNEL staining. (B) Quantification of TUNEL-positive cells in islets. n = 15 islets for each condition. *p < 0.05 vs. Nrf2+/+ islets with the same treatment. #p < 0.05 vs. Nrf2+/+ islets with iAs3+ treatment.
Nrf2 activation protects MIN6 cells against acute iAs3+ cytotoxicity in an Nrf2-dependent fashion
To provide further support of our hypothesis that Nrf2 activation protects pancreatic β-cells against iAs3+ cytotoxicity, the effect of pretreatment of MIN6 cells with Nrf2 activator tBHQ on iAs3+-induced cytotoxicity was examined. As shown in Fig. 6A, pretreatment of Scr cells with non-cytotoxic levels of tBHQ (50 µM) for 6 hrs showed a modest but statistically significant protection against subsequent iAs3+ toxicity. However, pretreatment of Nrf2-KD cells with tBHQ showed no protective effect (Fig. 6B), suggesting that the protective effect of tBHQ pretreatment against iAs3+ toxicity is a result of Nrf2 activation. Interestingly, NAC showed a significant protective effect from iAs3+ toxicity in Nrf2-KD cells, suggesting that exogenous antioxidants may rescue the impairment of the cellular antioxidant system caused by Nrf2 deficiency.
Fig. 6.
(A and B) Effect of pretreatment with non-cytotoxic levels of tBHQ on iAs3+ cytotoxicity in Scr (A) and Nrf2-KD (B) MIN6 cells. Cells were pretreated with 50 µM tBHQ for 6 hrs followed by a 24-hr iAs3+ exposure. (C and D) Effect of NAC on the cytotoxicity of iAs3+ in Scr (C) and Nrf2-KD (D) MIN6 cells. Cell viability was measured using the Non-Radioactive Cell-Proliferation Assay Kit. n = 6. *, p < 0.05 vs. Non-pretreated cells with the same concentration of iAs3+.
Discussion
Diabetes is a chronic disease characterized by high levels of glucose in the blood. There are four clinical classes, including Type 1 diabetes (T1D), T2D, gestational diabetes and other specific types of diabetes due to other causes. T1D results from autoimmune pancreatic β-cell destruction, usually leading to absolute insulin deficiency (Atkinson et al., 2011), whereas T2D results from a progressive insulin secretion defect on the background of insulin resistance (Robertson, 2006). While the precise mechanisms for the diabetogenic effect of iAs are still largely undefined, recent in vitro and in vivo experimental studies indicated that iAs or its metabolites impair insulin-dependent glucose uptake and result in insulin resistance (Izquierdo-Vega et al., 2006; Paul et al., 2007a; Paul et al., 2007b; Paul et al., 2011; Xue et al., 2011). In addition, accumulating epidemiological studies demonstrate that insulin resistance is a common symptom in diabetes associated with chronic iAs exposure (Lai et al., 1994; Rahman et al., 1998; Wang et al., 2003; Tseng, 2004; Nabi et al., 2005; Chiu et al., 2006; Coronado-Gonzalez et al., 2007; Meliker et al., 2007; Navas-Acien et al., 2008; Navas-Acien et al., 2009; Del Razo et al., 2011). Thus, T2D is the most predominant, if not only, type of diabetes associated with iAs exposure. It is clear that insulin resistance plays an early pathogenic role in the development of T2D and defects in insulin secretion by pancreatic β-cells are instrumental in the progression to hyperglycemia (Lowell and Shulman, 2005). In isolated rodent islets and cultured β-cells, iAs was found to decrease insulin transcription and GSIS (Diaz-Villasenor et al., 2006; Pi et al., 2007b; Fu et al., 2011). In the present study, we found that iAs3+ and CH3AsO, a form of MMA3+, induce apoptosis and/or cytotoxicity in β-cell line MIN6 cells and isolated mouse islets in a concentration- and time-dependent fashion. These findings suggest that β-cell damage and the associated dysfunction in insulin synthesis and secretion may be an important contributing factor for iAs-induced diabetes.
Although the precise molecular mechanisms of β-cell dysfunction in T2D are not completely understood, oxidative stress has been increasingly implicated in the pathogenesis of progressive β-cell failure (Evans et al., 2003; Kajimoto and Kaneto, 2004; Robertson and Harmon, 2006). It has been well documented that excessive and/or sustained increases in ROS production can directly or indirectly disturb the physiological function of cellular macromolecules such as DNA, protein or lipids and activate cellular stress-sensitive signaling pathways (Evans et al., 2003). In the early stages of oxidative stress, the adaptive response, primarily regulated by Nrf2, is the main response that will upregulate antioxidant and phase II detoxification enzymes and protect cells from more serious oxidative damage for cell survival. In the absence of an appropriate compensatory response from the endogenous antioxidant network, oxidative stress may cause oxidative damage and activate the cell death machinery (Davies, 2000). In this regard, abolishment of the Nrf2-mediated antioxidant response by targeted disruption of the Nrf2 gene in β-cell line MIN6 cells and mouse islets increased the susceptibility to iAs3+ and MMA3+-induced cytotoxicity and/or apoptosis. In contrast, activation of Nrf2 with tBHQ pretreatment significantly protects MIN6 cells from iAs3+-induced acute cytotoxicity in an Nrf2-dependent manner. These findings suggest that Nrf2 plays a critical role in pancreatic β-cell defense against iAs3+ and MMA3+-induced oxidative/electrophilic stress.
Although potentially cytotoxic, ROS also function as important intracellular signaling molecules for cellular responses to a variety of physiological stimuli, including glucose sensing in pancreatic β-cells (Pi et al., 2007b; Pi et al., 2010b). The involvement of ROS as signaling intermediates suggests that their magnitude would be inversely correlated with the ROS-scavenging activity and antioxidant status in the cell. Therefore, we propose that Nrf2-mediated antioxidant response plays a paradoxical role in arsenic-induced pancreatic β-cell dysfunction. On the one hand, it protects β-cells from high-dose arsenic-induced oxidative damage and possible cell death, thus minimizing oxidative damage-related impairment of insulin secretion. This premise was demonstrated by the present study indicating that Nrf2-mediated antioxidant response protects pancreatic β-cells from acute cytotoxicity of iAs3+ and MMA3+. On the other hand, because ROS signaling triggered by glucose could be an important component driving insulin secretion, the induction of endogenous antioxidants in the presence of iAs3+ and/or its methylated metabolites may blunt the ROS signaling resulting in reduced GSIS. Thus, we envision the following scenarios for arsenic-induced impairment of β-cell function. Under low, environmentally relevant levels of arsenic exposure, β-cells can adapt to the condition adequately by activating the Nrf2-ARE system, thereby keeping oxidative damage/cell death-related impairment of GSIS at a minimum. However, under chronic exposure conditions, the adaptively increased endogenous antioxidant capacity may interfere with glucose-dependent ROS signals that we postulate directly contribute to GSIS, leading reversibly to attenuated GSIS. Under high-dose arsenic exposure, the Nrf2 system may protect the cells from irreversible oxidative damage and possible cell death, which may be the primary cause for impaired GSIS under high-dose conditions. Thus the machinery of GSIS in β-cells is likely to be highly sensitive to arsenic exposure, though distinct mechanisms may apply through the dose continuum. These considerations are compatible with the view that oxidative stress may contribute to both early and late phases of β-cell failure in T2D.
In summary, the present study demonstrated that a deficiency in Nrf2-mediated antioxidant response renders pancreatic β-cells vulnerable to arsenic-induced cell damage, whereas activation of Nrf2 protects β-cells from arsenic toxicity. Considering the likely inhibitory effect of persistent Nrf2 activation on ROS signaling in GSIS, the potential application of activating Nrf2 to prevent and/or treat T2D associated with arsenic exposure should be approached with caution.
Supplementary Material
Highlights.
Lack of Nrf2 reduced expression of antioxidant genes induced by iAs3+ in β-cells.
Deficiency of Nrf2 in β-cells sensitized to iAs3+ and MMA3+-induced cytotoxicity
Nrf2 activation protected β-cells from acute iAs3+ cytotoxicity
Acknowledgements
This work was supported in part by the National Institutes of Health Grants DK76788 (to J.P.) and ES016005 (to J.P.). We are grateful for assistance from Dr. Masayuki Yamamoto at Tohoku University, Japan and Drs. Steve Kleeberger and Hye-Youn Cho at NIEHS in providing the Nrf2−/− mice. We also thank Lisa H. Webb, Kathy Bragg, Carol Bobbitt, Steve Butler and Paul Ross for their careful animal care and breeding management.
Abbreviations
- 18S
18S ribosomal RNA
- ARE
antioxidant response element
- DMA3+
dimethylarsinous acid
- DMEM
Dulbecco’s modified Eagle’s medium
- FBS
fetal bovine serum
- GCLC
γ-glutamate cysteine ligase catalytic subunit
- GSIS
glucose-stimulated insulin secretion
- Hmox1
heme oxygenase 1
- iAs
inorganic arsenic
- iAs3+
inorganic arsenite
- MMA3+
monomethylarsonous acid
- Nqo1
NAD(P)H: quinone oxidoreductase 1
- Nrf2
Nuclear factor E2-related factor 2
- Nrf2−/−
Nrf2-knockout
- Nrf2-KD
Nrf2-knockdown
- PBS
phosphate buffered saline
- PI
propidium iodide
- ROS
reactive oxygen species
- SFN
sulforaphane
- Srx
sulfiredoxin 1
- tBHQ
tert-Butylhydroquinone
- T1D
Type 1 diabetes
- T2D
Type 2 diabetes
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of interest
The content is solely the responsibility of the authors. All authors have agreed to its content. M.E.A received some funding from DOW Chemical Company and Unilever. All of the other authors declare no competing financial interests. B.Y., J.F., H.Z., P.X., K.Y., C.G.W, Y.H., Q.Z., M.E.A. and J.P. are employees of The Hamner Institutes for Health Sciences. The Hamner is a 501(c)3 non-profit organization that has a diverse research portfolio including funding from the American Chemistry Council, a trade association that represents chemical manufacturers.
References
- Aposhian HV. Enzymatic methylation of arsenic species and other new approaches to arsenic toxicity. Annu Rev Pharmacol Toxicol. 1997;37:397–419. doi: 10.1146/annurev.pharmtox.37.1.397. [DOI] [PubMed] [Google Scholar]
- Atkinson MA, Bluestone JA, Eisenbarth GS, Hebrok M, Herold KC, Accili D, Pietropaolo M, Arvan PR, Von Herrath M, Markel DS, Rhodes CJ. How does type 1 diabetes develop?: the notion of homicide or beta-cell suicide revisited. Diabetes. 2011;60:1370–1379. doi: 10.2337/db10-1797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bailey KA, Hester SD, Knapp GW, Owen RD, Thai SF. Gene expression of normal human epidermal keratinocytes modulated by trivalent arsenicals. Mol Carcinog. 2010;49:981–998. doi: 10.1002/mc.20677. [DOI] [PubMed] [Google Scholar]
- Chiu HF, Chang CC, Tsai SS, Yang CY. Does arsenic exposure increase the risk for diabetes mellitus? J Occup Environ Med. 2006;48:63–67. doi: 10.1097/01.jom.0000184854.75053.03. [DOI] [PubMed] [Google Scholar]
- Coronado-Gonzalez JA, Del Razo LM, Garcia-Vargas G, Sanmiguel-Salazar F, Escobedo-de la Pena J. Inorganic arsenic exposure and type 2 diabetes mellitus in Mexico. Environ Res. 2007;104:383–389. doi: 10.1016/j.envres.2007.03.004. [DOI] [PubMed] [Google Scholar]
- Cullen WR, McBride BC, Manji H, Pickett AW, Reglinski J. The metabolism of methylarsine oxide and sulfide. Appl. Organomet. Chem. 1989;3:71–78. [Google Scholar]
- Davies KJ. Oxidative stress, antioxidant defenses, and damage removal, repair, and replacement systems. IUBMB Life. 2000;50:279–289. doi: 10.1080/713803728. [DOI] [PubMed] [Google Scholar]
- Del Razo LM, Garcia-Vargas GG, Valenzuela OL, Castellanos EH, Sanchez-Pena LC, Currier JM, Drobna Z, Loomis D, Styblo M. Exposure to arsenic in drinking water is associated with increased prevalence of diabetes: a cross-sectional study in the Zimapan and Lagunera regions in Mexico. Environ Health. 2011;10:73. doi: 10.1186/1476-069X-10-73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Diaz-Villasenor A, Sanchez-Soto MC, Cebrian ME, Ostrosky-Wegman P, Hiriart M. Sodium arsenite impairs insulin secretion and transcription in pancreatic beta-cells. Toxicol Appl Pharmacol. 2006;214:30–34. doi: 10.1016/j.taap.2005.11.015. [DOI] [PubMed] [Google Scholar]
- Evans JL, Goldfine ID, Maddux BA, Grodsky GM. Are oxidative stress-activated signaling pathways mediators of insulin resistance and beta-cell dysfunction? Diabetes. 2003;52:1–8. doi: 10.2337/diabetes.52.1.1. [DOI] [PubMed] [Google Scholar]
- Fu J, Woods CG, Yehuda-Shnaidman E, Zhang Q, Wong V, Collins S, Sun G, Andersen ME, Pi J. Low-level arsenic impairs glucose-stimulated insulin secretion in pancreatic beta cells: involvement of cellular adaptive response to oxidative stress. Environ Health Perspect. 2011;118:864–870. doi: 10.1289/ehp.0901608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hayakawa T, Kobayashi Y, Cui X, Hirano S. A new metabolic pathway of arsenite: arsenic-glutathione complexes are substrates for human arsenic methyltransferase Cyt19. Arch Toxicol. 2005;79:183–191. doi: 10.1007/s00204-004-0620-x. [DOI] [PubMed] [Google Scholar]
- Itoh K, Chiba T, Takahashi S, Ishii T, Igarashi K, Katoh Y, Oyake T, Hayashi N, Satoh K, Hatayama I, Yamamoto M, Nabeshima Y. An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements. Biochem Biophys Res Commun. 1997;236:313–322. doi: 10.1006/bbrc.1997.6943. [DOI] [PubMed] [Google Scholar]
- Itoh K, Mimura J, Yamamoto M. Discovery of the negative regulator of Nrf2, Keap1: a historical overview. Antioxid Redox Signal. 2010;13:1665–1678. doi: 10.1089/ars.2010.3222. [DOI] [PubMed] [Google Scholar]
- Izquierdo-Vega JA, Soto CA, Sanchez-Pena LC, De Vizcaya-Ruiz A, Del Razo LM. Diabetogenic effects and pancreatic oxidative damage in rats subchronically exposed to arsenite. Toxicol Lett. 2006;160:135–142. doi: 10.1016/j.toxlet.2005.06.018. [DOI] [PubMed] [Google Scholar]
- Kajimoto Y, Kaneto H. Role of oxidative stress in pancreatic beta-cell dysfunction. Ann N Y Acad Sci. 2004;1011:168–176. doi: 10.1007/978-3-662-41088-2_17. [DOI] [PubMed] [Google Scholar]
- Lai MS, Hsueh YM, Chen CJ, Shyu MP, Chen SY, Kuo TL, Wu MM, Tai TY. Ingested inorganic arsenic and prevalence of diabetes mellitus. Am J Epidemiol. 1994;139:484–492. doi: 10.1093/oxfordjournals.aje.a117031. [DOI] [PubMed] [Google Scholar]
- Liu Q, Zhang H, Smeester L, Zou F, Kesic M, Jaspers I, Pi J, Fry RC. The NRF2-mediated oxidative stress response pathway is associated with tumor cell resistance to arsenic trioxide across the NCI-60 panel. BMC Med Genomics. 2010;3:37. doi: 10.1186/1755-8794-3-37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lowell BB, Shulman GI. Mitochondrial dysfunction and type 2 diabetes. Science. 2005;307:384–387. doi: 10.1126/science.1104343. [DOI] [PubMed] [Google Scholar]
- Maher J, Yamamoto M. The rise of antioxidant signaling--the evolution and hormetic actions of Nrf2. Toxicol Appl Pharmacol. 2010;244:4–15. doi: 10.1016/j.taap.2010.01.011. [DOI] [PubMed] [Google Scholar]
- Meliker JR, Wahl RL, Cameron LL, Nriagu JO. Arsenic in drinking water and cerebrovascular disease, diabetes mellitus, and kidney disease in Michigan: a standardized mortality ratio analysis. Environ Health. 2007;6:4. doi: 10.1186/1476-069X-6-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nabi AH, Rahman MM, Islam LN. Evaluation of Biochemical Changes in Chronic Arsenic Poisoning among Bangladeshi Patients. Int J Environ Res Public Health. 2005;2:385–393. doi: 10.3390/ijerph2005030002. [DOI] [PubMed] [Google Scholar]
- Navas-Acien A, Silbergeld EK, Pastor-Barriuso R, Guallar E. Arsenic exposure and prevalence of type 2 diabetes in US adults. JAMA. 2008;300:814–822. doi: 10.1001/jama.300.7.814. [DOI] [PubMed] [Google Scholar]
- Navas-Acien A, Silbergeld EK, Pastor-Barriuso R, Guallar E. Rejoinder: Arsenic exposure and prevalence of type 2 diabetes: updated findings from the National Health Nutrition and Examination Survey, 2003–2006. Epidemiology. 2009;20:816–820. doi: 10.1097/EDE.0b013e3181afef88. discussion e811-812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paul DS, Harmon AW, Devesa V, Thomas DJ, Styblo M. Molecular mechanisms of the diabetogenic effects of arsenic: inhibition of insulin signaling by arsenite and methylarsonous Acid. Environ Health Perspect. 2007a;115:734–742. doi: 10.1289/ehp.9867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paul DS, Hernandez-Zavala A, Walton FS, Adair BM, Dedina J, Matousek T, Styblo M. Examination of the effects of arsenic on glucose homeostasis in cell culture and animal studies: Development of a mouse model for arsenic-induced diabetes. Toxicol Appl Pharmacol. 2007b doi: 10.1016/j.taap.2007.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paul DS, Walton FS, Saunders RJ, Styblo M. Characterization of the impaired glucose homeostasis produced in C57BL/6 mice by chronic exposure to arsenic and high-fat diet. Environ Health Perspect. 2011;119:1104–1109. doi: 10.1289/ehp.1003324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Petrick JS, Jagadish B, Mash EA, Aposhian HV. Monomethylarsonous acid (MMA(III)) and arsenite: LD(50) in hamsters and in vitro inhibition of pyruvate dehydrogenase. Chem Res Toxicol. 2001;14:651–656. doi: 10.1021/tx000264z. [DOI] [PubMed] [Google Scholar]
- Pi J, Bai Y, Daniel KW, Liu D, Lyght O, Edelstein D, Brownlee M, Corkey BE, Collins S. Persistent oxidative stress due to absence of uncoupling protein 2 associated with impaired pancreatic beta-cell function. Endocrinology. 2009;150:3040–3048. doi: 10.1210/en.2008-1642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pi J, Bai Y, Reece JM, Williams J, Liu D, Freeman ML, Fahl WE, Shugar D, Liu J, Qu W, Collins S, Waalkes MP. Molecular mechanism of human Nrf2 activation and degradation: role of sequential phosphorylation by protein kinase CK2. Free Radic Biol Med. 2007a;42:1797–1806. doi: 10.1016/j.freeradbiomed.2007.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pi J, Bai Y, Zhang Q, Wong V, Floering LM, Daniel K, Reece JM, Deeney JT, Andersen ME, Corkey BE, Collins S. Reactive oxygen species as a signal in glucose-stimulated insulin secretion. Diabetes. 2007b;56:1783–1791. doi: 10.2337/db06-1601. [DOI] [PubMed] [Google Scholar]
- Pi J, Diwan BA, Sun Y, Liu J, Qu W, He Y, Styblo M, Waalkes MP. Arsenic-induced malignant transformation of human keratinocytes: involvement of Nrf2. Free Radic Biol Med. 2008;45:651–658. doi: 10.1016/j.freeradbiomed.2008.05.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pi J, He Y, Bortner C, Huang J, Liu J, Zhou T, Qu W, North SL, Kasprzak KS, Diwan BA, Chignell CF, Waalkes MP. Low level, long-term inorganic arsenite exposure causes generalized resistance to apoptosis in cultured human keratinocytes: Potential role in skin co-carcinogenesis. Int J Cancer. 2005;116:20–26. doi: 10.1002/ijc.20990. [DOI] [PubMed] [Google Scholar]
- Pi J, Horiguchi S, Sun Y, Nikaido M, Shimojo N, Hayashi T, Yamauchi H, Itoh K, Yamamoto M, Sun G, Waalkes MP, Kumagai Y. A potential mechanism for the impairment of nitric oxide formation caused by prolonged oral exposure to arsenate in rabbits. Free Radic Biol Med. 2003a;35:102–113. doi: 10.1016/s0891-5849(03)00269-7. [DOI] [PubMed] [Google Scholar]
- Pi J, Leung L, Xue P, Wang W, Hou Y, Liu D, Yehuda-Shnaidman E, Lee C, Lau J, Kurtz TW, Chan JY. Deficiency in the nuclear factor E2-related factor- 2 transcription factor results in impaired adipogenesis and protects against diet-induced obesity. J Biol Chem. 2010a;285:9292–9300. doi: 10.1074/jbc.M109.093955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pi J, Qu W, Reece JM, Kumagai Y, Waalkes MP. Transcription factor Nrf2 activation by inorganic arsenic in cultured keratinocytes: involvement of hydrogen peroxide. Exp Cell Res. 2003b;290:234–245. doi: 10.1016/s0014-4827(03)00341-0. [DOI] [PubMed] [Google Scholar]
- Pi J, Yamauchi H, Kumagai Y, Sun G, Yoshida T, Aikawa H, Hopenhayn-Rich C, Shimojo N. Evidence for induction of oxidative stress caused by chronic exposure of Chinese residents to arsenic contained in drinking water. Environ Health Perspect. 2002;110:331–336. doi: 10.1289/ehp.02110331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pi J, Zhang Q, Fu J, Woods CG, Hou Y, Corkey BE, Collins S, Andersen ME. ROS signaling, oxidative stress and Nrf2 in pancreatic beta-cell function. Toxicol Appl Pharmacol. 2010b;244:77–83. doi: 10.1016/j.taap.2009.05.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prentki M, Nolan CJ. Islet beta cell failure in type 2 diabetes. J Clin Invest. 2006;116:1802–1812. doi: 10.1172/JCI29103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rahman M, Tondel M, Ahmad SA, Axelson O. Diabetes mellitus associated with arsenic exposure in Bangladesh. Am J Epidemiol. 1998;148:198–203. doi: 10.1093/oxfordjournals.aje.a009624. [DOI] [PubMed] [Google Scholar]
- Robertson RP. Oxidative stress and impaired insulin secretion in type 2 diabetes. Curr Opin Pharmacol. 2006 doi: 10.1016/j.coph.2006.09.002. [DOI] [PubMed] [Google Scholar]
- Robertson RP, Harmon JS. Diabetes, glucose toxicity, and oxidative stress: A case of double jeopardy for the pancreatic islet beta cell. Free Radic Biol Med. 2006;41:177–184. doi: 10.1016/j.freeradbiomed.2005.04.030. [DOI] [PubMed] [Google Scholar]
- Tseng CH. The potential biological mechanisms of arsenic-induced diabetes mellitus. Toxicol Appl Pharmacol. 2004;197:67–83. doi: 10.1016/j.taap.2004.02.009. [DOI] [PubMed] [Google Scholar]
- Wang SL, Chiou JM, Chen CJ, Tseng CH, Chou WL, Wang CC, Wu TN, Chang LW. Prevalence of non-insulin-dependent diabetes mellitus and related vascular diseases in southwestern arseniasis-endemic and nonendemic areas in Taiwan. Environ Health Perspect. 2003;111:155–159. doi: 10.1289/ehp.5457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang XJ, Sun Z, Chen W, Li Y, Villeneuve NF, Zhang DD. Activation of Nrf2 by arsenite and monomethylarsonous acid is independent of Keap1-C151: enhanced Keap1-Cul3 interaction. Toxicol Appl Pharmacol. 2008;230:383–389. doi: 10.1016/j.taap.2008.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woods CG, Fu J, Xue P, Hou Y, Pluta LJ, Yang L, Zhang Q, Thomas RS, Andersen ME, Pi J. Dose-dependent transitions in Nrf2-mediated adaptive response and related stress responses to hypochlorous acid in mouse macrophages. Toxicol Appl Pharmacol. 2009;238:27–36. doi: 10.1016/j.taap.2009.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xue P, Hou Y, Zhang Q, Woods CG, Yarborough K, Liu H, Sun G, Andersen ME, Pi J. Prolonged inorganic arsenite exposure suppresses insulin-stimulated AKT S473 phosphorylation and glucose uptake in 3T3-L1 adipocytes: Involvement of the adaptive antioxidant response. Biochem Biophys Res Commun. 2011;407:360–365. doi: 10.1016/j.bbrc.2011.03.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao R, Hou Y, Xue P, Woods CG, Fu J, Feng B, Guan D, Sun G, Chan JY, Waalkes MP, Andersen ME, Pi J. Long isoforms of NRF1 contribute to arsenic-induced antioxidant response in human keratinocytes. Environ Health Perspect. 2011;119:56–62. doi: 10.1289/ehp.1002304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao R, Hou Y, Zhang Q, Woods CG, Xue P, Fu J, Yarborough K, Guan D, Andersen ME, Pi J. Cross-regulations among NRFs and KEAP1 and effects of their silencing on arsenic-induced antioxidant response and cytotoxicity in human keratinocytes. Environ Health Perspect. 2012;120:583–589. doi: 10.1289/ehp.1104580. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.






