Abstract
Chronic exposure to inorganic arsenic (iAs), a contaminant of water and food supplies, is associated with many adverse health effects. A notable feature of iAs metabolism is sequential methylation reactions which produce mono- and di-methylated arsenicals that can contain arsenic in either the trivalent (III) or pentavalent (V) valence states. Because methylated arsenicals containing trivalent arsenic are more potent toxicants than their pentavalent counterparts, the ability to distinguish between the +3 and +5 valence states is a crucial property for physiologically based pharmacokinetic (PBPK) models of arsenicals to possess if they are to be of use in risk assessment. Unfortunately, current analytic techniques for quantifying arsenicals in tissues disrupt the valence state; hence, pharmacokinetic studies in animals, used for model calibration, only reliably provide data on the sum of the +3 and +5 valence forms of a given metabolite. In this paper we show how mathematical modeling can be used to overcome this obstacle and present a PBPK model for the dimethylated metabolite of iAs, which exists as either dimethylarsinous acid, (CH3)2AsIIIOH (abbreviated DMAIII) or dimethylarsinic acid, (CH3)2AsV(O)OH (abbreviated DMAV). The model distinguishes these two forms and sets a lower bound on how much of an organ’s DMA burden is present in the more reactive and toxic trivalent valence state. We conjoin the PBPK model to a simple model for DMAIII-induced oxidative stress in liver and use this extended model to predict cytotoxicity in liver in response to the high oral dose of DMAV. The model incorporates mechanistic details derived from in vitro studies and is iteratively calibrated with lumped-valence-state PK data for intravenous or oral dosing with DMAV. Model formulation leads us to predict that orally administered DMAV undergoes extensive reduction in the gastrointestinal (GI) tract to the more toxic trivalent DMAIII.
Keywords: arsenic, trivalent, pentavalent
INTRODUCTION
Although best known for its role at acute doses as a metabolic poison, chronic sub-acute exposure to inorganic arsenic (iAs) increases the risk of skin, bladder, and lung cancers in humans (IARC, 2004), as well as the risk of diabetes and other pathologies (Abernathy et al., 2003). A common feature of arsenic metabolism in organisms as diverse as bacteria and humans is the conversion of inorganic arsenic (iAs) into methylated metabolites. In mammals, reactions that convert iAs into methylated metabolites are catalyzed by an S-adenosylmethionine-dependent arsenic methyltransferase, arsenic (+3 oxidation state) methyltransferase (As3mt) (Thomas and Rosen, 2013). The profile of arsenicals in urine from humans exposed to iAs reflects the extensive conversion of iAs to methylated metabolites: in a U.S. adult population exposed to iAs in drinking water, iAs accounted for 4.4 to 37.9% (1st and 99th percentiles), monomethylated arsenic for 5.9 to 29.3%, and dimethylated arsenic for 47.4 to 86.0% of the arsenic in urine (Hudgens et al., 2016).
In both inorganic and methylated species, arsenic can occur in a trivalent (AsIII) or pentavalent (AsV) state. Urine from individuals exposed to iAs contains arsenate (iAsV), arsenite (iAsIII), monomethylarsonic acid (MMAV), monomethylarsonous acid (MMAIII), dimethylarsinic acid (DMAV), and dimethylarsinous acid (DMAIII) (Aposhian et al., 2000; Le et al., 2000; Mandal et al., 2001; Wang et al., 2004; Valenzuela et al., 2005; Xie et al., 2006).
Trivalent methylated arsenicals have been found to be particularly toxic. In some cases, dimethylated arsenicals containing AsIII exceed both iAsIII and DMAV in potency as cytotoxins, genotoxins, and enzyme inhibitors (Styblo et al., 1997, 2000; Lin et al., 1999; Mass et al., 2001; Dopp et al., 2004, 2005; Naranmandura et al., 2011a, 2012; _Rehman et al., 2012a, b). In vitro studies have shown DMAIII to kill rat and human hepatocytes even when present in medium at concentrations less than 10 μM (Naranmandura et al., 2011b; Dopp et al., 2008), via the production of excessive levels of reactive oxygen species (ROS). In contrast, DMAV has been found to be non-cytotoxic even at 5000 μM (Dopp et al., 2008). Hence, it is crucial that a physiologically based pharmacokinetic (PBPK) model treat the two valence states separately. Unfortunately, experimental techniques available to quantify arsenicals in tissues disturb the valence state, limiting data available for model calibration.
In the current work, we show how mathematical modeling can be used to partially overcome this obstacle for DMA. We present a PBPK model in mouse which provides a lower bound on the concentration of DMAIII and an upper bound on the concentration of DMAV in liver, kidney, urinary bladder, and lung after oral or intravenous dosing with DMAIII or DMAV. Our model can be extended to pharmacodynamics applications. To illustrate this, we formulate a simple model for baseline and DMAIII-induced generation of hydrogen peroxide (H2O2), a major ROS, in hepatocytes, using the results of in vitro experiments investigating DMAIII cytotoxicity (Dopp et al., 2008). We then conjoin the H2O2 model to our PBPK model and predict H2O2 levels and cell death in mouse liver after the high oral dose of DMAV.
Although the model distinguishes DMA valence state, it was calibrated on lumped-valence-state PK data for arsenicals measured in liver, kidney, lung, urinary bladder, red blood cells, plasma, urine, and feces of mice that received DMAV intravenously (Hughes and Kenyon, 1998; Hughes et al., 2000) or orally (Hughes et al., 2008). We briefly discuss how our modeling approach differs from that taken by Evans et al., who also used these data sets to construct a PBPK model in mouse for orally and intravenously administered DMAV (Evans et al., 2008).
Evans et al. (2008) assumed no reduction of DMAV in any compartment. Hence, intravenously or orally administered DMAV remains in this valence form. A single compartment was used for the GI tract and it was assumed that all of an oral dose of DMAV is absorbed. First-order kinetics were used for all terms and biliary excretion to feces was neglected.
The data sets indicate that less than 5% of intravenously (IV) administered DMAV is excreted into feces, hence it is acceptable to neglect biliary excretion. However, the fecal data for oral gavage show a greater than 50% excretion of DMAV-derived species into feces, indicating incomplete absorption. Furthermore, the greater the oral dose, the larger the percentage found in feces, indicating saturable absorption kinetics.
Our model uses two compartments to model the GI tract. Doses begin in the first compartment and, if not absorbed, proceed to the second compartment; material not absorbed from the second compartment undergoes fecal excretion. Saturable (Michaelis-Menten) kinetics are used to describe absorption of DMAV from both GI compartments. The GI tract is modeled as it is using two compartments because orally ingested DMA is not immediately subject to fecal excretion, as it needs to progress through the GI tract.
Including fecal excretion led to the hypothesis, during fitting to the oral data sets, that a significant portion of orally administered DMAV undergoes reduction in the GI tract to the more toxic and rapidly absorbed trivalent form, DMAIII. Such reduction is likely in light of experimental work (Calatayud et al., 2013). We model this reduction using Michaelis-Menten kinetics, as this is reasonable for a process which may be catalyzed by gut bacteria (Rowland and Davies, 1981). Hence, our model predicts that an oral dose of DMAV results in both DMAV and DMAIII entering the bloodstream.
MATERIALS AND METHODS
Pharmacokinetic studies in mice
Three pharmacokinetic studies reported by Hughes and coworkers were used for model development and calibration. A 2000 study (Hughes et al., 2000) investigated the distribution of radiolabeled dimethylarsinic acid ([14C]-DMAV) administered intravenously to adult female B6C3F1 mice at two dosage levels (0.6 and 60 mg elemental As/kg). Mice were killed via cardiac puncture at intervals up to 2 hours post dosing. Concentration–time profiles of DMAV-derived radioactivity in liver, kidney, lung, red blood cells, and blood plasma were reported. Because neither methylated nor demethylated products of DMAV were detected in this study, tissue concentration-time course data represent solely dimethylated arsenicals.
A 1998 study (Hughes and Kenyon, 1998) using the same dosing protocol in adult female B6C3F1 mice reported cumulative percent dose of DMAV-derived radioactivity in urine at various time points over 24 hours post dosing. Because less than 5% of the dose was excreted in feces within 24 hours of dosing, we chose to neglect fecal excretion for intravenously administered DMAV in model construction.
Hughes et al. (2008) investigated the distribution of orally administered (gavage) radiolabeled dimethylarsinic acid ([14C]-DMAV) in adult female B6C3F1 mice at dosage levels of 0.6 and 60 mg elemental As/kg. Mice were housed in individual metabolism cages to collect urine and feces and killed by cardiac puncture at intervals up to 24 hours. Concentration–time profiles of DMAV-derived radioactivity in liver, kidney, lung, urinary bladder, and whole blood were reported, as were cumulative percent dose in urine and feces. In the same study, arsenic speciation analyses were performed in 24-hour urine collections from mice that received unlabeled DMAV at the 0.6 and 60 mg elemental As/kg dosage levels. For both dosage levels, urine contained DMAV, DMAIII, and trimethylarsine oxide (TMAO); TMAO is a trimethylated metabolite which is only produced in minor quantity in mice.
PBPK model for DMA
The PBPK model consists of ten compartments for both DMAV and DMAIII: a compartment representing the stomach and upper small intestine, a compartment representing the lower small intestine and the cecum, and compartments representing the blood plasma, the red blood cells, the liver, the kidneys, the lungs, the urinary bladder, the richly perfused tissues, and the slowly perfused tissues. In our model, the arterial and venous blood are combined. In addition to the compartments for DMAV and DMAIII, there is a plasma compartment for TMAO. A schematic of the PBPK model is shown in Figure 1 and model parameters are given in Tables 1 and 2.
Figure 1.
Schematic diagram for the DMAIII/DMAV PBPK model.
Table 1.
Physiological model parameters.
| Parameters | Values | References |
|---|---|---|
| Body weight, BW (kg) | 0.025 | mean weight in Hughes et al. experiments |
| Cardiac output, QCC (L/h/kg0.75) | 16.5 | Brown et al., 1997, p. 454 |
| Cardiac plasma output, QCPC (L/h/kg0.75) | 9.0750 | Equal to QCC(1-hem) |
| Hematocrit (hem) | 0.45 | Hedrich, 2004 |
| Blood flows (fraction of cardiac output, dimensionless) | ||
| Liver (Qliverf) | 0.161 | Brown et al., 1997, Table 23 |
| Kidneys (Qkidneyf) | 0.091 | Brown et al., 1997, Table 23 |
| Lungs (Qlungf) | 0.005 | Brown et al., 1997, Table 23 |
| Urinary bladder (QUBf) | 0.0033 | Stott et al., 1983, Table 1 |
| Richly perfused (QRf) | 0.4997 | Calculated to balance |
| Slowly perfused (QSf) | 0.24 | Arms & Travis, 1988, 4–35 |
| Tissue volumes (fraction of body weight, dimensionless) | ||
| Blood (Vbloodf) | 0.049 | Brown et al., 1997, Table 21 |
| Liver (Vliverf) | 0.055 | Brown et al., 1997, Table 21 |
| Kidneys (Vkidneyf) | 0.017 | Brown et al., 1997, Table 21 |
| Lungs (Vlungf) | 0.007 | Brown et al., 1997, Table 21 |
| Urinary bladder (VUBf) | 0.0009 | Stott et al., 1983, Table 2 |
| Richly perfused (VRf) | 0.0101 | Arms & Travis, 1988, 4–11 (sum of liver and richly perfused with liver, kidneys, lungs, and urinary bladder subtracted out) |
| Slowly perfused (VSf) | 0.771 | Calculated to balance after subtracting out 9% for skeletal tissue; close to Arms & Travis, 1988, 4–11 |
| Capillary volumes in tissues (fraction of tissue volume, dimensionless) | ||
| Fraction of liver volume that is blood (Vcapliverf) | 0.31 | Brown et al., 1997, Table 30 |
| Fraction of kidney volume that is blood (Vcapkidneyf) | 0.24 | Brown et al., 1997, Table 30 |
| Fraction of lung volume that is blood (Vcaplungf) | 0.5 | Brown et al., 1997, Table 30 |
| Fraction of urinary bladder volume that is blood (VcapUBf) | 0.03 | Brown et al., 1997, Table 30 (assumed equal to adrenal) |
| Fraction of slowly perfused volume that is blood (VcapSf) | 0.03 | Brown et al., 1997, Table 30 (assumed equal to skin) |
| Fraction of richly perfused volume that is blood (VcapRf) | 0.31 | assumed equal to liver |
Table 2.
Chemical-specific model parameters (see text). Tissue-to-plasma distribution ratios for DMAV are consistent with Dong et al. (2016).
| Parameters | Values |
|---|---|
| Tissue-to-plasma distribution ratios for DMAV (unitless) | |
| Liver (Pliver5) | 1 |
| Kidneys (Pkidney5) | 6 |
| Lungs (Plung5) | 2 |
| Urinary bladder (PUB5) | 3 |
| Richly perfused tissues (PR5) | 2 |
| Slowly perfused tissues (PS5) | 5 |
| Tissue-to-plasma distribution ratios for DMAIII (unitless) | |
| Liver (Pliver3) | 100 |
| Kidneys (Pkidney3) | 600 |
| Lungs (Plung3) | 200 |
| Urinary bladder (PUB3) | 300 |
| Richly perfused tissues (PR3) | 200 |
| Slowly perfused tissues (PS3) | 500 |
| Permeability coefficients for DMAV (L/h/kg0.75) | |
| Liver (πliverC5) | 0.02 |
| Kidneys (πkidneyC5) | 0.2 |
| Lungs (πlungC5) | 0.001 |
| Urinary bladder (πUBC5) | 0.001 |
| Richly perfused tissues (πRC5) | 0.1 |
| Slowly perfused tissues (πSC5) | 0.3 |
| Permeability coefficients for DMAIII (L/h/kg0.75) | |
| Liver (πliverC3) | 0.25 |
| Kidneys (πkidneyC3) | 2 |
| Lungs (πlungC3) | 0.5 |
| Urinary bladder (πUBC3) | 0.01 |
| Richly perfused tissues (πRC3) | 1 |
| Slowly perfused tissues (πSC3) | 3 |
| Oral dosing parameters | |
| Scaling factor for maximal rate of absorption of DMAV from stomach/upper small intestine and lower small intestine/cecum compartments (Vmax,abs5C) (μmol/h/kg0.75) | 50 |
| Scaling factor for Km for absorption of DMAV from stomach/upper small intestine and lower small intestine/cecum compartments (Km,abs5C) (μmol/kg) | 2000 |
| Scaling factor for maximal rate of reduction of DMAV to DMAIII in stomach/upper small intestine and lower small intestine/cecum compartments (Vmax,redC) (μmol/h/kg0.75) | 120 |
| Scaling factor for Km for reduction of DMAV to DMAIII in stomach/upper small intestine and lower small intestine/cecum compartments (Km,redC) (μmol/kg) | 800 |
| First-order rate constant for absorption of DMAIII from stomach/upper small intestine and lower small intestine/cecum compartments (kabs3C) (h−1·kg0.25) | 1 |
| First-order rate constant for methylation of DMAIII in lower small intestine/cecum compartment to TMAO appearing in plasma (k3→TMAOC) (h−1·kg0.25) | 1 |
| First-order rate constant for passage of DMAV and DMAIII from stomach/upper small intestine compartment to lower small intestine/cecum compartment (ktoGI2C) (h−1·kg0.25) | 0.25 |
| First-order rate constant for removal of DMAV and DMAIII from lower small intestine/cecum compartment to feces (ktofecesC) (h−1·kg0.25) | 0.4 |
| First-order rate constants for oxidation of DMAIII to DMAV in tissues (L/h/kg0.75) | |
| Liver (kliver3→5C) | 0.1 |
| Kidneys (kkidney3→5C) | 0.03 |
| Lungs (klung3→5C) | 0.01 |
| Urinary bladder (kUB3→5C) | 0.002 |
| Richly perfused tissues (kR3→5C) | 0.02 |
| Slowly perfused tissues (kS3→5C) | 2 |
| Red blood cells (kRBC3→5C) | 0.04 |
| First-order rate constants for exchange of DMAV and DMAIII between red blood cells and plasma (L/h/kg0.75) | |
| Uptake of DMAV in plasma by red blood cells (kplasmatorbc5C) | 0.008 |
| Release of DMAV in red blood cells to plasma (krbctoplasma5C) | 0.02 |
| Uptake of DMAIII in plasma by red blood cells (kplasmatorbc3C) | 0.2 |
| Release of DMAIII in red blood cells to plasma (krbctoplasma3C) | 0.03 |
| First-order rate constants for urinary elimination (L/h/kg0.75) | |
| Excretion of TMAO from plasma to urine (kTMAO,urineC) | 0.3 |
| Excretion of DMAIII from kidney capillaries to urine (kDMA3,urineC) | 1 |
| Excretion of DMAV from kidney capillaries to urine (kDMA5,urineC) | 1 |
For the two compartments modeling the gastrointestinal (GI) tract, the PBPK model equations specify the time rate of change in the amount of DMAV or DMAIII in the compartment (units of μmol per hour). For all other compartments, the equations specify the time rate of change in the concentrations (μM per hour). All rate equations are statements of mass balance. The rate of increase in the amount or concentration of a chemical in a compartment (or capillary or cellular portion of a compartment, as described later) is the sum of the rates at which it is taken up by the compartment and produced within it, less the sum of the rates at which it is released or lost internally by conversion to another species. Model equations were coded in MATLAB and solutions computed using the ode15s solver (MATLAB R2015b, The MathWorks Inc., Natick, MA, 2015).
The model was developed for the mouse and parameterized to enable allometric scaling to rats or humans. Hence, in the model equations which follow, where a parameter x appears in an equation but Tables 1 or 2 only indicate a parameter xC, xC must be scaled by the organism body weight (0.025 kg for the mouse) to yield the parameter value x featured in the equation.
Compartments modeling the stomach/upper small intestine and lower small intestine/cecum
DMAV administered orally begins in the compartment modeling the stomach/upper small intestine, where it (1) is absorbed at a rate governed by Michaelis-Menten kinetics, (2) undergoes reduction to DMAIII at a rate governed by Michaelis-Menten kinetics, and (3) is transferred from this compartment to the lower small intestine/cecum compartment by a process with first-order kinetics. In the lower small intestine/cecum compartment, we assume that DMAV not only continues to be absorbed and reduced at the same rates as in the stomach/upper small intestine compartment, but is also excreted into feces.
Our choice of kinetics for DMAV absorption is based on two observations of the mouse data sets. First, less than 5% of intravenously administered DMAV was excreted in feces, indicating that most DMAV found in feces after oral administration was non-absorbed material. Second, fecal excretion of orally administered DMAV showed dose dependency, with a higher fractional excretion at the higher dose level (Hughes et al., 2008). This behavior can be modeled using saturating (Michaelis-Menten) absorption kinetics. We obtained by optimization an effective Km and Vmax for DMAV absorption across the intestinal epithelium. We emphasize that here we take a phenomenological approach and do not make mechanistic claims. There is experimental evidence that the multidrug resistance-associated protein 4 (MRP4) functions in DMAV efflux across the basolateral membrane of enterocytes (Roggenbeck et al., 2016), but there is insufficient data to model this transporter explicitly. A similar approach underlies our handling of DMAIII, as little is known about epithelial transporters involved in the movement of this species (Roggenbeck et al., 2016).
Bacteria in the rat gastrointestinal (GI) tract have been found to reduce arsenate (AsV) to arsenite (AsIII) (Rowland and Davies, 1981), suggesting that a similar pathway may exist for DMAV reduction. Studies in in vitro systems that mimic the functioning of the GI tract indicate that DMAV is reduced to DMAIII (Calatayud et al., 2013). Notably, the in vitro system replicated the pH changes associated with digestive processes but lacked the microbiota of the GI tract. In light of these findings, we assume some reduction of DMAV to DMAIII in the GI tract, as discussed below.
DMAIII appears via reduction of DMAV in the compartment modeling the stomach/upper small intestine, where it (1) is absorbed with first-order kinetics and (2) enters the lower small intestine/cecum compartment with the same kinetics as DMAV (i.e., first-order kinetics with the same rate constant). DMAIII in the lower small intestine/cecum compartment (1) is absorbed at the same rate as in the compartment modeling the stomach/upper small intestine, (2) undergoes methylation to TMAO with first-order kinetics, and (3) is excreted into feces with the same kinetics as DMAV (first-order kinetics with the same rate constant). Evidence exists for the methylation of DMAV to TMAO by the bacteria of mouse cecum (Kubachka et al., 2009). This is likely a two-step process in which reduction of DMAV to trivalency precedes methylation. For simplicity in modeling, we do not track TMAO in the lower small intestine/cecum compartment but rather have it appear directly in the blood plasma. Therefore, the associated rate constant is a lumped parameter representing both methylation and absorption.
The model equations for the two GI tract compartments are given below. GI15 and GI13 are the quantities (in μmol) of DMAV and DMAIII, respectively, in the compartment modeling the stomach/upper small intestine. GI25 and GI23 are the quantities of DMAV and DMAIII in the compartment modeling the lower small intestine/cecum. Parameter definitions and values are given in Table 2. Notably, the two Km values associated with absorption and reduction of DMAV actually have units of amount (μmol) rather than concentration. This discrepancy reflects the lack of volume estimates for the two compartments modeling the GI tract, as these compartments are phenomenological, rather than physiologically defined. Thus, the two Km values scale linearly with body volume (body weight) rather than remain constant, with scaling constants given by Km,redC and Km,abs5C (see Table 2).
| (1) |
| (2) |
| (3) |
| (4) |
Liver, kidneys, lungs, urinary bladder, and slowly and richly perfused compartments
Because DMAV is poorly membrane-permeable (Dopp et al., 2004, 2005; Naranmandura et al., 2011a), we model its pharmacokinetics as diffusion-limited rather than flow-limited. This requires us to model separately the cellular and capillary portions of each organ compartment (including the slowly and richly perfused compartments), and to have a separate equation for each dictating the time rate of change in DMAV concentration. Cellular uptake of DMAIII is much more rapid (Dopp et al., 2004, 2005; Naranmandura et al., 2011a) and in the absence of oxidation, flow-limited kinetics would probably be appropriate. However, Hippler et al. (2011) found that MMAIII taken up by cells is rapidly oxidized to MMAV, likely by lysosomal degradation of MMAIII-containing complexes. We assume the same process could oxidize DMAIII in cells. We model the intracellular oxidation of DMAIII to DMAV with a first-order term, with rate constant k3→5. We do not model a reduction of DMAV inside cells because there is little information on the quantitative significance of this reaction. By neglecting reduction, we create a model that provides a lower bound on the concentration of DMAIII and an upper bound on the concentration of DMAV in an organ. For ease of coupling the equations for DMAIII with those for DMAV, we also use diffusion-limited kinetics for DMAIII.
DMAV and DMAIII enter the capillary portion of an organ from the plasma compartment (a lumped arterial/venous compartment) and exit via venous drainage, with exchange occurring between the capillary and cellular portions of that organ. For a given organ with volume V=BW∙Vf, where BW is body weight (0.025 kg for the mouse) and Vf is the fraction of body weight constituted by the organ, the volume of the capillary portion is Vcap=V·Vcapf and the volume of the cellular portion is Vcell=V·(1-Vcapf). Within the capillaries, only species dissolved in the plasma (rather than associated with the red blood cells) can be immediately taken up by the cells of an organ. Hence, it is actually the capillary plasma concentration that is modeled. We model the plasma portion of the capillaries as having volume (1-hem)Vcap, where hem is the hematocrit value (0.45 for mouse). The plasma flow Q into an organ is Q=Qf∙QCP, where Qf is the fraction of cardiac output received by the organ and QCP is the plasma portion of the cardiac output, related to the whole-blood cardiac output QC via QCP =QC(1-hem).
With these considerations in mind, the liver, kidneys, urinary bladder, lungs, and slowly and richly perfused compartments each have four equations of the form given below governing the time rates of change in the concentrations of DMAIII and DMAV, with the following organ-specific modifications: the terms describing loss of DMAIII and DMAV from the two GI tract compartments to systemic absorption appear as additional input terms to the liver capillary equations, and the kidney capillary equations feature additional terms describing loss of DMAIII and DMAV to urine. We model the urinary loss using first-order kinetics and the same rate constant for both valence states.
| (5) |
| (6) |
| (7) |
| (8) |
Cap5 and Cell5 are the concentrations of DMAV in the capillary plasma and cellular portions of an organ, respectively; Cap3 and Cell3 are the concentrations of DMAIII in the capillary plasma and cellular portions of an organ, respectively. Pl5 and Pl3 are the concentrations of DMAV and DMAIII in the plasma compartment. Permeability coefficients are denoted by π and partition coefficients by P, with subscript indicating valence state. We note that, with the exception of hem, the parameters featured in the above equations are different for each organ compartment (see Tables 1 and 2).
TMAO is present in very small quantities and is taken up even more slowly than DMAV (Dopp et al., 2004); hence, we do not model its uptake or release by organs. Rather, TMAO created in the lower small intestine/cecum compartment appears directly in the blood plasma, where it is subject to urinary loss (see equations in the next section). Unlike the rat enzyme, mouse liver As3mt does not efficiently metabolize DMAIII to TMAO (Fomenko et al., 2007) and we neglect this reaction in our model.
Plasma and red blood cell compartments
The equations governing arsenical concentrations in the blood plasma and red blood cells are given below. RBC3 and RBC5 are the concentrations of DMAIII and DMAV, respectively, in the red blood cell compartment. TMAO is the concentration of trimethylarsine oxide in the plasma compartment.
| (9) |
| (10) |
| (11) |
| (12) |
| (13) |
The volume of the plasma compartment is given by (1-hem)Vblood and that of the red blood cells by hem∙Vblood, where Vblood is the volume of whole blood. DMAV and DMAIII leaving organ capillaries via venous drainage enter the plasma compartment (a lumped venous/arterial compartment). DMAV and DMAIII leave the plasma compartment and enter organ capillaries via arterial flow. We model diffusion of DMAV and DMAIII between plasma and red blood cells. In addition, oxidation of DMAIII to DMAV in red blood cells (Shiobara et al., 2001) is modeled. TMAO created in the lower small intestine/cecum compartment appears in the blood plasma and is subject to urinary excretion. Due to its slow cellular uptake and lack of pharmacokinetic data, we do not model its cellular uptake or exchange with red blood cells.
Parameterization and calibration
Values and references for the physiological parameters are given in Table 1. Chemical-specific parameter values were determined by fitting of model solutions to the PK data sets, under the assumption of mouse weight 0.025 kg. We term this fitting process “model calibration.” Chemical-specific parameter values are given in Table 2.
In the mouse studies, levels of DMAV-derived species in organs were reported as μg elemental As per liter. In our model, concentrations are computed in units of μmol per liter. Therefore, model solutions for concentrations in organs were scaled by 75 g/mol, the molecular weight of elemental arsenic. To determine the concentration of DMAV-derived species in an organ for comparison with experimental data, we summed the amounts of DMAV and DMAIII in the cellular and capillary portions of the organ and divided by the total organ volume. That is, we did not assume that all arsenicals measured to be in an organ were intracellular, but that some portion of the total organ As burden resides in the capillaries. This approach is reasonable for a chemical with diffusion-limited kinetics, such as DMAV, and was consistent with the use of non-perfused mouse tissues for arsenical analysis.
In the mouse studies, DMAV-derived species measured in cumulative urine and feces at various time points were expressed as percent administered dose. We converted these data to units of μg elemental As, using average mouse body weight of 0.025 kg and administered dosage levels of 0.6 or 60 mg As/kg. In our model, DMAV-derived species (DMAV, DMAIII, and TMAO) in cumulative urine and feces are computed in units of μmol; these were scaled by the molecular weight of elemental As for comparison with experimental data. In the urine speciation portion of the mouse studies, concentrations of DMAV, DMAIII, and TMAO in urine were reported in units of μg As per ml. For comparison with model output, we scaled these data points by the average 24-hour urinary output of a mouse of 1.6 ml (Green, 1966).
In determining chemical-specific parameter values by optimization, we adhered to the following constraints, informed by in vitro studies. First, for each organ, the permeability coefficient for DMAIII was required to be at least an order of magnitude greater than that for DMAV. This is consistent with studies of cellular uptake of trivalent and pentavalent arsenicals (Dopp et al., 2004). Second, for each organ, the distribution ratio for DMAIII was required to be approximately 100 times greater than that for DMAV. 99% of intracellular iAsIII is bound to sulfhydryl groups (Kitchin, 2011) and if we assume a similar figure holds for DMAIII, only 1% of DMAIII in a cell would be available for release at any given time. This assumption results in a DMAIII distribution ratio that is 100 times greater than that for DMAV, which is assumed to be mainly free. Third, the rate of absorption kabs3 of DMAIII from the GI compartments was required to be an order of magnitude greater than Vmax,abs5/Km,abs5, consistent with in vitro studies aimed at characterizing the intestinal absorption of DMAIII and DMAV (Calatayud et al., 2010; Calatayud et al., 2011).
The model was calibrated iteratively. In the first step, permeability coefficients and distribution ratios for DMAV in the compartments, rate constants for the diffusion of DMAV between plasma and red blood cells, and the urinary excretion rate constant for DMAV were determined by fitting to the intravenous data sets. We found that an excellent fit of the intravenous data sets could be obtained by assuming no reduction of DMAV occurs outside of the gastrointestinal tract, thus reducing the intravenous case to a DMAV-only submodel. There is a paucity of reliable data on the relative quantities of DMAIII and DMAV reached at equilibrium inside of cells, and making this assumption yields a useful model in that the model provides a lower bound on the DMAIII concentration and an upper bound on the DMAV concentration in an organ. After obtaining these parameters, remaining parameters were obtained through fitting to the oral data sets.
Sensitivity analysis
MATLAB code was written to perform a local sensitivity analysis. For each chemical-specific model parameter, the normalized sensitivity coefficient (NSC) was calculated using the following equation, while all other parameters remained at their original values.
| (14) |
Here, P is the original parameter value, O is the model output resulting from the original parameter value, Pi is the parameter value increased by 1%, and Mi is the model output resulting from the new parameter value. For bolus dosing protocols, such as those considered here, model outputs are not at steady state; hence, the NSC for each parameter is actually a function of time. Parameters with NSCs exceeding 0.1 in absolute value in some compartment at some time point are classified as sensitive.
In performing the sensitivity analysis for the DMAV distribution ratios, DMAV permeability coefficients, rate constants for the diffusion of DMAV between plasma and red blood cells, and the urinary excretion rate constant for DMAV, the model outputs considered were the concentration-time courses in plasma, red blood cells, lungs, liver, kidneys, and cumulative amount in urine of total DMAV-derived species resulting from intravenous dosing with the low and high doses of DMAV. An important function of a sensitivity analysis is to determine which data would be most helpful in identifying the value of a given model parameter (e.g., concentration at a given time point in a given organ). This process is as effective as the model is accurate; hence, we chose to do the sensitivity analysis for these parameters using only intravenous dosing with DMAV as this approach effectively reduces the PBPK model to a DMAV-only submodel, minimizing inaccuracy caused by errors in the model’s handling of DMAIII as well as its absorption from gut.
For the remaining parameters, the model outputs used for the sensitivity analysis were the concentration-time courses in whole blood, liver, kidneys, urinary bladder, lungs, and cumulative amount in urine of total DMAV-derived species resulting from oral dosing with the high and low doses of DMAV. Because these parameters have no impact on model output for intravenously administered DMAV, only oral doses were used for this part of the sensitivity analysis.
Pharmacodynamic application: DMAIII-induced oxidative stress
We model the baseline production and removal of hydrogen peroxide in liver cells with a simple linear equation, with parameters Vh2o2prod and kh2o2. Sies (2014) gives a value of 50 nnol/min·g tissue for the rate of production of H2O2 in liver. We assume that the liver is 70% water by weight and convert Sies’s value to units of μM/hr to obtain a value of 4286 μM/hr for Vh2o2prod. Sies states that the normal concentration of H2O2 in liver is 0.01 μM, and kh2o2 is chosen to yield this value for the steady-state concentration. The presence of DMAIII is assumed to increase the rate of H2O2 production above baseline, with the strength of the effect depending upon a parameter dmaprod. Our equation for H2O2 is given below.
| (15) |
To determine the parameter value dmaprod, we created a model of an in vitro system studied by Dopp et al. (2008) in which human hepatocytes were exposed to DMAIII in medium and the concentration causing 50% cell death within 24 hours determined, referred to as the LC 50 dose; analogous studies for mouse have not been done, to the best of our knowledge. To model this system, we conjoined Equation (15) with five additional equations, four modeling DMAIII and DMAV pharmacokinetics, and one modeling hepatocyte survival. Hepatocytes are assumed in the model to have a death rate which scales linearly with H2O2 concentration. The equations are given below.
| (16) |
| (17) |
| (18) |
| (19) |
| (20) |
Parameter values and descriptions are given in Table 3. Partition coefficients remain the same as in the PBPK model, but permeability coefficients as well as the rate constant for the oxidation of DMAIII to DMAV are scaled by the ratio of vcell to the mouse liver tissue volume for the PBPK model. The value of the parameter death was chosen by holding H2O2 concentration constant at 3000 μM and finding a parameter value which results in 80% cell death after 4 hours (Surv=0.2), consistent with experiments conducted by Kanno et al. (1999) investigating the ability of H2O2 to induce cell death. Finally, dmaprod was chosen so that an initial DMAIII concentration in medium of 8.6 μM results in Surv=0.5 after 24 hours (Dopp et al., 2008). With the value of dmaprod determined, equations (15) and (20) were appended to the DMA PBPK mouse model and the impact on liver of the high oral dose of DMAV investigated.
Table 3.
In vitro model parameters (see text).
| Parameter | Description | Value |
|---|---|---|
| π3 | Permeability coefficient for DMAIII | 1.29429e-5 L/h |
| π5 | Permeability coefficient for DMAV | 1.03544e-6 L/h |
| k3→5 | Rate constant for oxidation of DMAIII to DMAV | 5.17718e-6 L/h |
| vcell | Volume of cells in culture | 0.78125e-6 L |
| vmed | Volume of medium | 499.21875e-6 L |
| death | Hepatocyte death rate | 0.00013 μM−1h−1 |
| dmaprod | Parameter governing increase in rate of H2O2 production over baseline, due to DMAIII | 1500 μM−1 |
| Vh2o2prod | Baseline rate of production of H2O2 in hepatocytes | 4.286e3 μM h−1 |
| kh2o2 | Rate constant for H2O2 removal | 4.286e5 h−1 |
RESULTS
Calibration data
Using the chemical-specific parameter values given in Table 2, model solutions were computed for intravenous and oral dosing of a 0.025 kg mouse with 0.6 and 60 mg As/kg [14C]-DMAV. Figure 2 compares the total radioactivity in urine, kidneys, liver, lungs, blood plasma, and red blood cells as computed by the model, for intravenous dosing, with experimental data. Model solutions fall within a factor of 2 of most data points, taking into account error bars. Slightly more disagreement occurs for the early data points in plasma, especially for the lower dose. Here, total radioactivity in plasma computed by the model at the 0.25-hr mark is about 1/6 the observed value.
Figure 2.
Model solutions compared with experimental data (Hughes and Kenyon, 1998; Hughes et al., 2000) for intravenous dosing with [14C]-DMAV. Total DMAV-derived radioactivity is given in units of μ elemental As (in cumulative urine, Figure 2A) and micrograms elemental As per liter (all other graphs). High dose is 60 mg As/kg, low dose is 0.6 mg As/kg. Legend: low dose model solution (dashed line), low dose experimental data (solid dots), high dose model solution (solid line), high dose experimental data (asterisks).
Figure 3 compares model solutions for total radioactivity in urine, feces, kidneys, liver, lungs, urinary bladder, blood, and 24-hr cumulative urine speciation with experimental data from the oral dosing studies. Model solutions fall within a factor of 2 of most data points. For the lower dose, total radioactivity in urinary bladder at the 8-hr mark and kidneys at the 4-hr mark as computed by the model are both equal to about 0.25 the observed values. Early time points in feces disagree significantly with the model. However, we note that what the model actually computes is the quantity of DMAIII/V destined for fecal excretion at any given time. Given the time required for intestinal transit before excretion, this discrepancy between observed and predicted values is not unexpected.
Figure 3.
Model solutions compared with experimental data collected by Hughes et al. (2008) for oral dosing with [14C]-DMAV (total radioactivity graphs, Fig. 3A-3G) or with unlabeled DMAV (urine speciation graphs, Fig. 3H-3J). High dose is 60 mg As/kg, low dose is 0.6 mg As/kg. Legend: low dose model solution (dashed line), low dose experimental data (solid dots), high dose model solution (solid line), high dose experimental data (asterisks).
The fitting process provided insight into the fate of DMAV in the GI tract. Michaelis-Menten kinetics were assumed for both the absorption of DMAV into the bloodstream and its reduction in the GI tract to DMAIII. We determined that the reductive process is favored, having both a higher capacity (higher Vmax) and higher affinity (lower Km) than the absorptive process. This result indicates that orally ingested DMAV is extensively reduced in the GI tract to the more toxic trivalent species before being absorbed. If we do not assume extensive reduction of DMAV in the GI tract, then model solutions greatly under-predict experimental data in all organs. The assumption of no reduction is equivalent, in terms of model structure, to simplifying our approach by using a single state variable which represents the sum of both DMAIII and DMAV. Figure 4 shows that making the trivalent/pentavalent distinction is crucial for obtaining a good fit to experimental data in liver; the situation in liver was representative of all organs.
Figure 4.
Model predictions for total radioactivity in liver after the high oral dose of DMAV (60 mg As/kg). Fig. 4A was generated from a model using a single state variable to represent the sum of the +3 and +5 valence states. Fig. 4B was generated using our full model which separately tracks DMAIII and DMAV and assumes reduction of DMAV to DMAIII in the GI tract.
Valence-state specific tissue dosimetry predictions
A prime objective of the model is to make separate tissue dosimetry predictions for DMAV and DMAIII. Figure 5 shows the predicted minimal amount of DMAIII and maximal amount of DMAV in the cells of lung, urinary bladder, liver, and kidney for the larger oral dose of DMAV (60 mg As/kg). We predict that lung cells could encounter a DMAV concentration as high as 35,000 μg/L, while kidney cells are predicted to encounter a concentration of DMAIII exceeding 12,000 μg/L.
Figure 5.
Model predictions for DMAV (solid line) and DMAIII (dashed line) in cellular portion of tissues (not including capillary bed) after the high oral dose of DMAV (60 mg As/kg).
For each organ, the peak concentration for DMAIII occurs earlier than that for DMAV. This temporal difference reflects the model assumptions that DMAIII has a higher permeability coefficient than DMAV (hence, is taken up more rapidly) and is oxidized to DMAV once inside the cell. The organs differ in their ratios of DMAIII to DMAV, due to differences in permeability coefficients, distribution ratios, rates of blood flow through the organs, and organ volumes. For comparison, Fig. 6 shows predicted tissue dosimetry in liver and lung cells when the same dose of DMAV is administered intravenously. Our model assumes no reduction of DMAV outside of the GI tract, hence DMAIII concentration is zero. Oral and intravenous administration are predicted to yield a similar peak concentration of DMA (sum of DMAIII and DMAV) in liver despite the fact that, in oral dosing, less than half as much DMAV enters the system, the rest being excreted into feces. This is due to the much greater permeability of DMAIII versus DMAV. In lung, even a larger peak concentration of DMA is achieved for oral versus IV administration. Furthermore, with oral dosing significant quantities of the more toxic DMAIII are produced. Hence, our model predicts that the oral route is significantly more toxic than the IV route.
Figure 6.
Model predictions for DMAV (solid line) and DMAIII (dashed line) in cellular portion of liver (Fig. 6A) and lung (Fig. 6B) after intravenous injection with the high oral dose of DMAV (60 mg As/kg).
Compare with Figures 5C and 5A, respectively, where the same dose was given orally.
Sensitivity analysis
For each chemical-specific model parameter, normalized sensitivity coefficients (as a function of time) were computed in response to either oral or intravenous dosing with DMAV (see Methods), where the model outputs considered were total DMAV-derived species in organ compartments and cumulative urine. When intravenous dosing was used for a given parameter, NSCs were computed for red blood cells, blood plasma, lungs, liver, kidneys, and urine. When oral dosing was used, NSCs were computed for blood, liver, kidneys, lungs, urinary bladder, and urine.
Sensitivity analyses found that the urinary excretion rate constant for DMAV and the permeability coefficients and distribution ratios for DMAV in liver, kidneys, and lungs were all sensitive, with NSCs exceeding 0.1 in magnitude for significant stretches of time in one or more compartments. Figure 7 provides NSCs for liver. The two parameters governing diffusion of DMAV between red blood cells and plasma were very sensitive, with NSCs in the red blood cell compartment near 1 in magnitude for large time intervals falling within the first two hours after dosing.
Figure 7.
Normalized sensitivity coefficients in the liver compartment for the permeability coefficient (Fig. 7A) and distribution ratio (Fig. 7B) of DMAV in liver. Computed for intravenous dosing with DMAV (60 mg As/kg).
Sensitivity analyses found that the two parameters governing diffusion of DMAIII between red blood cells and plasma and the permeability coefficients for DMAIII in all organs except lung were sensitive. Oral dosing enabled us to identify the permeability coefficient and distribution ratio for DMAV in urinary bladder (an organ for which concentration-time data were not reported for intravenous dosing): both these parameters had NSCs exceeding 0.1 in the urinary bladder compartment for most of the first 24 hours following oral dosing. None of the distribution ratios for DMAIII was sensitive. Parameters governing the rate of oxidation of DMAIII to DMAV were only sensitive in kidney and liver. All parameters associated with the GI tract were sensitive, as were the rate constants for the urinary elimination of DMAIII and TMAO.
LC 50 of DMAIII in hepatocytes
The parameter governing excess H2O2 production induced by intracellular DMAIII (dmaprod, see Table 3) was chosen so that the in vitro model predicts the death of approximately half of hepatocytes in culture after 24 hours in response to an initial DMAIII concentration in medium of 8.6 μM, consistent with Dopp et al. (2008). Fig. 8 shows the intracellular DMAIII concentration, intracellular H2O2 concentration, and fraction of surviving cells computed by the in vitro model.
Figure 8.
Intracellular DMAIII concentration (left panel), intracellular H2O2 concentration (middle panel), and fraction of surviving cells (right panel) computed by the in vitro model for an initial DMAIII concentration in medium of 8.6 μM.
Predicted oxidative stress and cytotoxicity in mouse liver after the high oral dose of DMAV
After determining dmaprod, Equations (15) and (20) were inserted into the DMA PBPK model, enabling us to make in vivo predictions of oxidative stress in liver and resultant liver injury in response to dosing with DMA. We predict that the high oral dose of DMAV (60 mg As/kg) causes intracellular H2O2 levels to exceed 1000 μM (normal concentration is 0.01 μM) and causes the death of about 1/3 of the liver after 6 hours; see Fig. 9. Fig. 5C shows the predicted intracellular DMAIII concentration, assumed to be the causative agent. Studies of hepatectomy in mouse have determined that the liver can regenerate even after 70% removal (Hori et al., 2011). Hence, our extended PBPK/PD model is consistent with the fact that this was not a lethal dose, although it does predict significant liver cell death. We note that our value for dmaprod was determined using data on DMAIII toxicity in human hepatocytes. Model accuracy could be improved if analogous data for mouse hepatocytes became available.
Figure 9.
Intracellular H2O2 concentration in liver (Fig. 9A) and fraction of surviving hepatocytes (Fig. 9B) computed by the PBPK/PD model for the high oral dose of DMAV (60 mg As/kg).
DISCUSSION
Model solutions for the sum of DMAIII and DMAV in tissues are in excellent agreement with lumped-valence data on the distribution of arsenicals in mice after oral or intravenous dosing with DMAV that was collected by Hughes and coworkers. Model solutions fall within a factor of two of most data points, taking into account error bars. One of the reasons we were able to achieve this level of agreement was our decision to posit that DMAV-derived species in an organ include not only intracellular arsenicals but also arsenicals present in residual blood in the organ’s capillary bed. The assumption that arsenicals retained in an organ could be present in the capillary bed is appropriate because organs were not perfused to remove residual blood from the capillaries and DMAV has diffusion-limited kinetics, unlike the small lipophilic molecules more typically dealt with in PBPK modeling. Hence, the model output that is compared with experimental data is the concentration of DMAV-derived species averaged over the cellular and capillary portions of an organ.
Model formulation led to the prediction that there is extensive reduction of DMAV to DMAIII in the GI tract. In our model, DMAV can be reduced to DMAIII in both GI compartments, and one possible fate for the DMAIII so produced is absorption. Hence, some of an oral dose of DMAV is ultimately taken up by tissues in the form of DMAIII, for which the permeability coefficients are larger. We find that if we do not assume such reduction, the model greatly under-predicts the oral dosing PK data in all organs. Extensive reduction of DMAV in the GI tract constitutes a model prediction which requires testing. It is very reasonable in light of the fact that cellular uptake of DMAV is quite slow (Dopp et al., 2004, 2005; Naranmandura et al., 2011a) and yet DMAV administered in water or the diet is rapidly excreted in urine. Although simulated digestion experiments (Calatayud et al., 2013) support the idea of a reduction of DMAV in gut, additional in vitro experiments are needed, preferably ones in which the simulated digestion system not only emulates the pH changes associated with digestion, but also includes gut microbiota.
In vitro studies with different cell types to determine permeability coefficients in various organs for DMAIII and DMAV, analogous to those conducted by Calatayud et al. (2010, 2011) for the study of intestinal transport, would do much to reduce model uncertainty. Although our values for DMAV distribution ratios in organs were arrived at through optimization, the values we found are consistent with empirically measured partition coefficients determined in two cases of fatal human poisoning (Dong et al., 2016). This consistency supports the idea that over time most DMAIII in the body undergoes oxidation to DMAV, and also lends support to our distribution ratio values for DMAV. Regarding the interconversion of the two valence forms of DMA within cells, we assumed that the rapid intracellular oxidation of MMAIII to MMAV reported by Hippler et al. (2011) also occurs for DMAIII. There are insufficient data to assess the quantitative significance of the reverse reaction, the reduction of DMAV to DMAIII. Therefore, reduction was omitted, yielding a model which provides a lower bound on the DMAIII concentration and an upper bound on the DMAV concentration inside cells.
We illustrated that our PBPK mouse model can be extended to pharmacodynamics applications, by conjoining it to a simple model for DMAIII-induced oxidative stress and resultant cytotoxicity in hepatocytes. Our prediction that the high oral dose of DMAV (60 mg As/kg) caused the death of 1/3 of the liver is consistent with the facts that this was not a lethal dose and that the liver can regenerate even after 70% hepatectomy (Hori et al., 2011).
To summarize, we have created the first PBPK model for a methylated arsenical which makes separate tissue dosimetry predictions for its +3 and +5 valence forms, models systemic circulation of both valence forms, and accommodates dosing with both valence forms. This ability is crucial because trivalent methylated arsenicals are now believed to play a large role in toxicity resulting from chronic exposure to iAs. Creating a valence-state distinguishing PBPK model for an arsenical is not a straightforward task because current analytic techniques for quantifying arsenicals in tissues disturb the valence state, creating a shortage of data for model calibration. Nevertheless, by making reasonable assumptions about where reduction of DMAV occurs in the body and utilizing the findings of in vitro studies we constructed a model which provides a lower bound on the concentration of DMAIII and an upper bound on the concentration of DMAV in tissues of interest.
Acknowledgments
FUNDING
This work was supported by the National Toxicology Program (NTP) and funded by an Interagency Agreement between the Food and Drug Administration [FDA IAG 224–12-0003] and the National Institute of Environmental Health Sciences at the National Institutes of Health [NIEHS IAG AES12013]. Lydia Bilinsky acknowledges the support of a fellowship from the Oak Ridge Institute for Science and Education, administered through an Interagency Agreement between the U.S. Department of Energy and the U.S. Food and Drug Administration. The views presented in this article do not necessarily reflect those of the U.S. Food and Drug Administration or National Toxicology Program. Preparation of this document has been funded, in part, by the U.S. Environmental Protection Agency (EPA). This document has been subjected to review by the National Health and Environmental Effects Research Laboratory (NHEERL), EPA and approved for publication. Approval does not signify that the contents reflect the views of the EPA, nor does mention of trade names or commercial products constitute endorsement or recommendation for use.
REFERENCES
- Abernathy CO, Thomas DJ, and Calderon RL (2003). Health effects and risk assessment of arsenic. The Journal of Nutrition. 133, 1536S–1538S. [DOI] [PubMed] [Google Scholar]
- Aposhian HV, Gurzau ES, Le XC, Gurzau A, Healy SM, Lu X, Ma M, Yip L, Zakharyan RA, Maiorino RM, Dart RC, Tircus MG, Gonzalez-Ramirez D, Morgan DL, Avram D, and Aposhian MM (2000). Occurrence of monomethylarsonous acid in urine of humans exposed to inorganic arsenic. Chem. Res. Toxicol 13, 693–697. [DOI] [PubMed] [Google Scholar]
- Arms AD and Travis CC Reference Physiological Parameters in Pharmacokinetic Modeling. Washington, D.C.: U.S. Environmental Protection Agency; 1988. [Google Scholar]
- Brown RP, Delp MD, Lindstedt SL, Rhomberg LR, and Beliles RP (1997). Physiological parameter values for physiologically based pharmacokinetic models. Toxicol. Ind. Health. 13, 407–484. [DOI] [PubMed] [Google Scholar]
- Calatayud M, Gimeno J, Velez D, Devesa V, and Montoro R (2010). Characterization of the intestinal absorption of arsenate, monomethylarsonic acid, and dimethylarsinic acid using the Caco-2 cell line. Chem. Res. Toxicol 23, 547–556. [DOI] [PubMed] [Google Scholar]
- Calatayud M, Devesa V, Montoro R, and Velez D (2011). In vitro study of intestinal transport of arsenite, monomethylarsonous acid, and dimethylarsinous acid by Caco-2 cell line. Toxicology Letters. 204, 127–133. [DOI] [PubMed] [Google Scholar]
- Calatayud M, Bralatei E, Feldmann J, Devesa V, and Velez D (2013). Transformation of arsenic species during in vitro gastrointestinal digestion of vegetables. Journal of agricultural and food chemistry. 61, 12164–12170. [DOI] [PubMed] [Google Scholar]
- Challenger F (1945). Biological methylation. Chem. Rev 36, 315–362. [Google Scholar]
- Dheeman DS, Packianathan C, Pillai JK, and Rosen BP (2014). Pathway of human AS3MT arsenic methylation. Chemical Research in Toxicology. 27, 1979–1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dong Z, Liu C, Liu Y, Yan K, Semple KT, and Naidu R (2016). Using publicly available data, a physiologically-based pharmacokinetic model and Bayesian simulation to improve arsenic non-cancer dose-response. Environment Intl 92-93, 239–46. [DOI] [PubMed] [Google Scholar]
- Dopp E, Hartmann LM, Florea AM, von Recklinghausen U, Pieper R, Shokouhi B, Rettenmeier AW, Hirner AV, and Obe G (2004). Uptake of inorganic and organic derivatives of arsenic associated with induced cytotoxic and genotoxic effect in Chinese hamster ovary (CHO) cells. Toxicology and Applied Pharmacology. 201,156–165. [DOI] [PubMed] [Google Scholar]
- Dopp E, Hartmann LM, von Recklinghausen U, Florea AM, Rabieh S, Zimmermann U, Shokouhi B, Yadav S, Hirner AV, and Rettenmeier AW (2005). Forced uptake of trivalent and pentavalent methylated and inorganic arsenic and its cyto-/genotoxicity in fibroblasts and hepatoma cells. Toxicological Sciences. 87, 46–56. [DOI] [PubMed] [Google Scholar]
- Dopp E, von Recklinghausen U, Hartmann LM, Stueckradt I, Pollok I, Rabieh S, Hao L, Nussler A, Katier C, Hirner AV, and Rettenmeier AW (2008). Subcellular distribution of inorganic and methylated arsenic compounds in human urothelial cells and human hepatocytes. Drug Metabolism and Disposition. 36, 971–979. [DOI] [PubMed] [Google Scholar]
- El-Masri HA, and Kenyon EM (2008). Development of a human physiologically based pharmacokinetic (PBPK) model for inorganic arsenic and its mono- and di-methylated metabolites. J Pharmacokinet Pharmacodyn. 35, 31–68. [DOI] [PubMed] [Google Scholar]
- Evans MV, Dowd SM, Kenyon EM, Hughes MF, and El-Masri HA (2008). A physiologically based pharmacokinetic model for intravenous and ingested dimethylarsinic acid in mice. Toxicological Sciences. 104, 250–260. [DOI] [PubMed] [Google Scholar]
- Fomenko DE, Xing W, Adair BM, Thomas DJ, and Gladyshev VN (2007). High-throughput identification of catalytic redox-active cysteine residues. Science. 315, 387–389. [DOI] [PubMed] [Google Scholar]
- Green EL, Ed. Biology of the Laboratory Mouse. New York: Dover Publications; 1966. [Google Scholar]
- Hayakawa T, Kobayashi Y, Cui X, and Hirano S (2005). A new metabolic pathway of arsenite: arsenic-glutathione complexes are substrates for human arsenic methyltransferase Cyt19. Archives of Toxicology. 79, 183–191. [DOI] [PubMed] [Google Scholar]
- Hippler J, Zdrenka R, Reichel RAD, Weber DG, Rozynek P, Johnen G, Dopp E, and Hirner AV (2011). Intracellular, time-resolved speciation and quantification of arsenic compounds in human urothelial and hepatoma cells. Journal of Analytical Atomic Spectrometry. 26, 2396–2403. [Google Scholar]
- Hori T, Ohashi N, Chen F, Baine AT, Gardner LB, Jermanus S, and Nguyen JH (2011). Simple and sure methodology for massive hepatectomy in the mouse. Ann.Gastroenterol. 24, 307–318. [PMC free article] [PubMed] [Google Scholar]
- Hudgens EE, Drobna Z, He B, Le XC, Styblo M, Rogers J, and Thomas DJ (2016).Biological and behavioral factors modify urinary arsenic metabolic profiles in a U.S. population. Environ. Health 15, 62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hughes MF and Kenyon EM (1998). Dose-dependent effects on the disposition of monomethylarsonic acid and dimethylarsinic acid in the mouse after intravenous administration. Journal of Toxicology and Environmental Health, Part A. 53, 95–112. [DOI] [PubMed] [Google Scholar]
- Hughes MF, Del Razo LM, and Kenyon EM (2000). Dose-dependent effects on tissue distribution and metabolism of dimethylarsinic acid in the mouse after intravenous administration. Toxicology. 143, 155–166. [DOI] [PubMed] [Google Scholar]
- Hughes MF, Devesa V, Adair BM, Conklin SD, Creed JT, Styblo M, Kenyon EM, and Thomas DJ (2008). Tissue dosimetry, metabolism and excretion of pentavalent and trivalent dimethylated arsenic in mice after oral administration. Toxicology and Applied Pharmacology. 227, 26–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- International Agency for Research on Cancer. IARC monographs on the evaluation of carcinogenic risks to humans, volume 84 Some drinking-water disinfectants and contaminants, including arsenic. Lyon, France: IARC; 2004. [PMC free article] [PubMed] [Google Scholar]
- Kanno S, Ishikawa M, Takayanagi M, Takayanagi Y, and Sasaki K (1999). Exposure to hydrogen peroxide induces cell death via apoptosis in primary cultured mouse hepatocytes. Biol. Pharm. Bull 22, 1296–1300. [DOI] [PubMed] [Google Scholar]
- Kitchin KT (2011). Arsenic’s interactions with macromolecules and its relationship to carcinogenesis In Biological Chemistry of Arsenic, Antimony and Bismuth (Sun H, Ed.), p. 22 John Wiley & Sons Ltd, Chichester. [Google Scholar]
- Kubachka KM, Kohan MC, Conklin SD, Herbin-Davis K, Creed JT, and Thomas DJ (2009). In vitro biotransformation of dimethylarsinic acid and trimethylarsine oxide by anaerobic microflora of mouse cecum analyzed by HPLC-ICP-MS and HPLC-ESI-MS. J. Anal. At. Spectrom 24, 1062–1068. [Google Scholar]
- Le XC, Ma M, Cullen WR, Aposhian HV, Lu X, and Zheng B (2000). Determination of monomethylarsonous acid, a key arsenic methylation intermediate, in human urine. Environ. Health Perspect 108, 1015–1018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin S, Cullen WR, and Thomas DJ (1999). Methylarsenicals and arsinothiols are potent inhibitors of mouse liver thioredoxin reductase. Chem Res Toxicol. 12, 924–930. [DOI] [PubMed] [Google Scholar]
- Mandal BK, Ogra Y, and Suzuki KT (2001). Identification of dimethylarsinous and monomethylarsonous acids in human urine of the arsenic-affected areas in West Bengal, India. Chem. Res. Toxicol 14, 371–378. [DOI] [PubMed] [Google Scholar]
- Mass MJ, Tennant A, Roop BC, Cullen WR, Styblo M, Thomas DJ, and Kligerman AD (2001). Methylated trivalent arsenic species are genotoxic. Chem. Res. Toxicol 14, 355–361. [DOI] [PubMed] [Google Scholar]
- Naranmandura H, Carew MW, Xu S, Lee J, Leslie EM, Weinfeld M, and Le XC (2011a). Comparative toxicity of arsenic metabolites in human bladder cancer EJ-1 cells. Chem. Res. Toxicol 24, 1586–1596. [DOI] [PubMed] [Google Scholar]
- Naranmandura H, Xu S, Sawata T, Hao WH, Liu H, Bu N, Ogra Y, Lou YJ, and Suzuki N (2011b). Mitochondria are the main target organelle for trivalent monomethylarsonous acid (MMAIII)-induced cytotoxicity. Chem. Res. Toxicol 24, 1094–1103. [DOI] [PubMed] [Google Scholar]
- Naranmandura H, Chen X, Tanaka M, Wang WW, Rehman K, Xu S, Chen Z, Chen SQ, and Suzuki N (2012). Release of apoptotic cytochrome C from mitochondria by dimethylarsinous acid occurs through interaction with voltage-dependent anion channel in vitro. Toxicol. Sci 128, 137–146. [DOI] [PubMed] [Google Scholar]
- Rehman K, Chen Z, Wang WW, Wang YW, Sakamoto A, Zhang YF, Naranmandura H, and Suzuki N (2012a). Mechanisms underlying the inhibitory effects of arsenic compounds on protein tyrosine phosphatase (PTP). Toxicol. Appl. Pharmacol 263, 273–280. [DOI] [PubMed] [Google Scholar]
- Rehman K, and Naranmandura H (2012b). Arsenic metabolism and thioarsenicals. Metallomics. 4, 881–892. [DOI] [PubMed] [Google Scholar]
- Roggenbeck BA, Banerjee M, and Leslie EM (2016). Cellular arsenic transport pathways in mammals. Journal of Environmental Sciences. 49, 38–58. [DOI] [PubMed] [Google Scholar]
- Rowland IR and Davies MJ (1981). In vitro metabolism of inorganic arsenic by the gastro-intestinal microflora of the rat. Journal of Applied Toxicology. 1, 278–283. [DOI] [PubMed] [Google Scholar]
- Sies H (2014). Role of metabolic H2O2 generation: redox signaling and oxidative stress. J. Biol. Chem 289, 8735–8741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shiobara Y, Ogra Y, and Suzuki KT (2001). Animal species difference in the uptake of dimethylarsinous acid (DMAIII) by red blood cells. Chemical Research in Toxicology. 14, 1446–1452. [DOI] [PubMed] [Google Scholar]
- Stott WT, Dryzga MD, and Ramsey JC (1983). Blood-flow distribution in the mouse. J. Appl. Toxicol 3, 310–312. [DOI] [PubMed] [Google Scholar]
- Styblo M, Serves SV, Cullen WR, and Thomas DJ (1997). Comparative inhibition of yeast glutathione reductase by arsenicals and arsenothiols. Chem. Res. Toxicol 10, 27–33. [DOI] [PubMed] [Google Scholar]
- Styblo M, Del Razo LM, Vega L, Germolec DR, LeCluyse EL, Hamilton GA, Reed W, Wang C, Cullen WR, and Thomas DJ (2000). Comparative toxicity of trivalent and pentavalent inorganic and methylated arsenicals in rat and human cells. Arch. Toxicol 74, 289–299. [DOI] [PubMed] [Google Scholar]
- Thomas DJ and Rosen BP (2013). Arsenic methyltransferase. In Encyclopedia of Metalloproteins (Kretsinger RH, Uversky VN, and Permyako EA, Eds.), pp. 138–143. Springer, Berlin. [Google Scholar]
- Valenzuela OL, Borja-Aburto VH, Garcia-Vargas GG, Cruz-Gonzalez MB, Garcia-Montalvo EA, Calderon-Aranda ES, and Del Razo LM (2005). Urinary trivalent methylated arsenic species in a population chronically exposed to inorganic arsenic. Environ. Health Perspect 113, 250–254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Z, Zhou J, Lu X, Gong Z, and Le XC (2004). Arsenic speciation in urine from acute promyelocytic leukemia patients undergoing arsenic trioxide treatment. Chem. Res. Toxicol 17, 95–103. [DOI] [PubMed] [Google Scholar]
- Xie R, Johnson W, Spayd S, Hall GS, and Buckley B (2006). Arsenic speciation analysis of human urine using ion exchange chromatography coupled to inductively coupled plasma mass spectrometry. Anal. Chim. Acta 578, 186–194 [DOI] [PubMed] [Google Scholar]










