{alpha}2u-Globulin Nephropathy, Renal Cell Proliferation, and Dosimetry of Inhaled tert-Butyl Alcohol in Male and Female F-344 Rats

Susan J. Borghoff1,, Judith S. Prescott, Derek B. Janszen, Brian A. Wong and Jeffery I. Everitt

CIIT Centers for Health Research, PO Box 12137, Six Davis Drive, Research Triangle Park, North Carolina 27709-2137

Received November 29, 2000; accepted January 8, 2001


    ABSTRACT
 TOP
 ABSTRACT
 INTRODUCTION
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
tert-Butyl alcohol (TBA) has been shown to cause kidney tumors in male rats following chronic administration in drinking water. The objective of the present study was to determine whether TBA induces {alpha}2u-globulin ({alpha}2u) nephropathy ({alpha}2u-N) and enhanced renal cell proliferation in male, but not female, F-344 rats, and whether the dosimetry of TBA to the kidney is gender specific. Male and female F-344 rats were exposed to 0, 250, 450, or 1750 ppm TBA vapors 6 h/day for 10 consecutive days to assess {alpha}2u-nephropathy and renal cell proliferation and for 1 and 8 days to evaluate the dosimetry of TBA following a single and repeated exposure scenario. Protein droplet accumulation was observed in kidneys of male rats exposed to 1750 ppm TBA, with {alpha}2u-globulin immunoreactivity present in these protein droplets. A statistically significant increase in {alpha}2u concentration in the kidney, as measured by an enzyme-linked immunosorbent assay, was observed in male rats exposed to 1750 ppm TBA with a exposure-related increase in renal cell proliferation. Renal {alpha}2u concentration was positively correlated with cell proliferation in male rat kidney. No histological lesions or increased renal cell proliferation was observed in female rats exposed to TBA compared to controls. The TBA kidney:blood ratio was higher at all concentrations and time points in male rats compared with female rats, which suggests that TBA is retained longer in male rat kidney compared with female rat kidney. Together these data suggest that TBA causes {alpha}2u-N in male rats, which is responsible for the male rat-specific increase in renal cell proliferation.

Key Words: tert-butyl alcohol (TBA); {alpha}2u-globulin ({alpha}2u)..


    INTRODUCTION
 TOP
 ABSTRACT
 INTRODUCTION
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
tert-Butyl alcohol (TBA) is a small aliphatic alcohol used in the manufacture of perfumes and cosmetics (Cosmetic Ingredient Review Expert Panel, 1989Go). TBA is also an indirect food additive when used in the preparation and application of coatings for paper and paperboard used in food containers (NTP, 1995Go). TBA is used in the production of isobutylene and in the manufacturing of methyl tert-butyl ether (MTBE), a commonly used fuel additive. Although TBA has been approved for use as an octane enhancer in unleaded gasoline, the use of TBA as a gasoline additive has not been widespread (U.S. EPA, 1991aGo).

TBA is a metabolite of MTBE in both rodents and humans. The pharmacokinetics of TBA following MTBE exposure are similar in these species; TBA accumulates in blood and is slowly cleared from blood (Amberg et al., 1999Go; Miller et al., 1997Go; Nihlen et al., 1998Go). In 13C-TBA–dosed rats, 13C-acetone, TBA, and its glucuronide were found to be minor metabolites and 2-methyl-1,2-propanediol and 2-hydroxyisobutyrate were the major metabolites present in urine, demonstrating that TBA is oxidatively metabolized or directly conjugated in vivo (Bernauer et al., 1998Go).

In a 2-year cancer study, administration of various levels of TBA in drinking water significantly increased the incidence of renal tumors in male F-344 rats and follicular cell adenomas of the thyroid in female mice (Cirvello et al., 1995Go; NTP, 1995Go). TBA did not induce any mutations when tested in four different strains of Salmonella typhimurium (Zeiger et al., 1987Go). More recently, TBA was shown to cause an increase in mutations in the TA102 strain of Salmonella following 2–4 mg of TBA/plate (Williams-Hill et al., 1999Go), but was negative in L5178Y mouse lymphoma cell assay with and without metabolic activation (McGregor et al., 1988Go). Overall, TBA is considered to cause tumors in rodents through a nongenotoxic mechanism.

A number of nongenotoxic chemicals that cause male rat-specific renal tumors also induce a syndrome unique to male rats, referred to as protein droplet, or {alpha}2u-globulin ({alpha}2u), nephropathy ({alpha}2u-N) (U.S. EPA, 1991bGo). More recently, MTBE has been added to this list (Bird et al., 1997Go; Prescott-Mathews et al., 1999Go). The proposed mechanism for {alpha}2u-N is dependent on the accumulation of the male rat–specific, low-molecular-weight protein {alpha}2u within the renal proximal tubule. Following glomerular filtration of {alpha}2u, approximately 50% of the protein is reabsorbed by proximal tubule cells, primarily in the P2 segment, while the remainder is excreted in the urine (Neuhaus et al., 1981Go). Under normal conditions, reabsorbed {alpha}2u is degraded by proteolytic enzymes within the phagolysosomes. Following exposure to {alpha}2u inducers, however, the parent compound or a metabolite binds {alpha}2u in a noncovalent, reversible manner, rendering {alpha}2u resistant to proteolytic hydrolysis. Massive accumulation of {alpha}2u overwhelms the phagolysosome, resulting in proximal tubule necrosis and compensatory cell proliferation (see review by Swenberg and Lehman-McKeeman, 1999). A sustained increase in cell proliferation is capable of increasing the likelihood of converting a spontaneous or chemically mediated error in DNA replication to a mutation and may enhance the clonal expansion of the initiated cell population to an eventual tumorigenic response (Butterworth et al., 1992Go). {alpha}2u-N is a male rat–specific response and does not occur in female rats or other species, including mice, guinea pigs, hamsters, dogs, and nonhuman primates (reviewed by Borghoff et al., 1990). Additionally, no evidence to date supports the synthesis of {alpha}2u or the development of {alpha}2u-N in humans (Borghoff and Lagarde, 1993Go; Lehman McKeeman and Caudill 1992Go; Olson et al., 1990Go).

In a 90-day TBA drinking water study, direct effects of TBA were found only in the kidney, ureter, and urinary bladder of exposed F-344 rats (Lindamood et al., 1992Go; Takahashi et al., 1993Go). Hyaline (protein) droplet deposition and hyaline crystals in tubular epithelium were enhanced in all male rats exposed to TBA, with the exception of the high-dose group (4% TBA). Enhanced cell proliferation, indicated by an increase in proliferating cell nuclear antigen (PCNA)–positive S-phase cells, was evident in the male rats exposed to 1% and 2% TBA. These changes correlated with the deposition of protein in the kidneys at the same concentrations (Lindamood et al., 1992Go; Takahashi et al., 1993Go). Additionally, histopathologic examination of hematoxylin-eosin (H&E)-stained male rat kidney sections revealed a dose-related increase in the severity of renal lesions, characterized by tubular atrophy, tubular regeneration, or thickening of the basement membranes in male rats administered 0.25, 0.5, 1.0, and 2.0% TBA, but not 4% TBA.

Although the TBA drinking water studies indicate that TBA induces protein droplet nephropathy in male F-344 rats, no investigation thus far has definitively demonstrated an exposure-related accumulation of {alpha}2u or specific renal cell proliferation in the male rats. Therefore, the objective of the present study was to determine whether TBA induces {alpha}2u-N and enhanced renal cell proliferation in male, but not female, F-344 rats, and if the dosimetry of TBA to the kidney is gender specific. Because water consumption was decreased for a considerable period of time in rats exposed to TBA in their water compared to controls (Lindamood et al., 1992Go), we elected to expose rats to TBA via inhalation. Based on review of archival H&E-stained kidney sections from an 18-day TBA inhalation study, exposure concentrations of 0, 250, 450, and 1750 ppm TBA were selected for the present investigation (NTP, 1997Go). To evaluate sex-specific retention of TBA, the disposition of TBA to the kidney of male and female rats exposed to TBA either for 1 day or 8 days via inhalation was also carried out. As {alpha}2u-N is a syndrome unique to male rats, elucidation of the mechanism of TBA-induced renal carcinogenesis is important not only in assessing the relevance of this response to humans, but also in understanding the role of TBA in the MTBE-induced renal tumor response in male rats.


    MATERIALS AND METHODS
 TOP
 ABSTRACT
 INTRODUCTION
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
Animals.
Rats were utilized in accordance with federal guidelines for the humane care and use of laboratory animals (NIH, 1985Go), and the study was approved by the Institutional Animal Care and Use Committee of the Chemical Industry Institute of Toxicology (CIIT). Male and female F-344 rats (CDF[F-344]/CrlBR) were obtained from Charles River Laboratories (Raleigh, NC) at 10 weeks of age and acclimated to the inhalation racks for 10 days prior to experimentation. During periods between exposures, animals were provided an NIH-07 pelleted diet (Zeigler Bros., Gardners, PA) and deionized, filter-purified water ad libitum.

Animal exposures.
Male and female rats were randomized (n = five rats/sex/concentration) prior to exposure. Animals were housed and exposed to TBA vapors for 6 h/day in 1-m3 H1000 stainless steel and glass chambers (H1000, Lab Products, Inc., Maywood, NJ). The H1000 chambers were contained within permanent 8-m3 Hinners-type chambers. The 8-m3 chambers served as an additional safety containment system. Temperature was maintained between 20–26°C and relative humidity between 40–60%. Air flow through the chamber ranged from 200 to 250 l/min, which maintained 12–15 air changes/h.

The exposure atmospheres of TBA (> 99% pure, CAS No. 75-65-0, Mallinckrodt, Raleigh, NC) were generated by metering known amounts of TBA with a fluid metering pump (Fluid Metering Inc., Oyster Bay, NY) into a stainless steel J-tube. The reservoir containing the TBA was immersed in a water bath maintained between 34 and 38°C to prevent the TBA from solidifying. Nitrogen was passed through the J-tube to carry the TBA vapor into the air supply of a 1-m3 exposure chamber. Prior to the start of exposures, uniformity of distribution of TBA in each exposure chamber was checked by measuring the concentration at nine positions (eight corners and the center of the chamber). The variation of concentration within the chamber was < 2% for all chambers.

The concentration of the test atmosphere in each chamber was analyzed approximately three time/h using a gas chromatograph (GC) (Hewlett Packard 5890 series II) equipped with a multiport sampling valve, a 0.5-ml sample loop, a flame ionization detector, and a 5-ft x 1/8" OD stainless steel column packed with 1.5% OV-101 on Chromasorb G-HP (Alltech, Deerfield, IL). Target concentrations of 0, 250, 450, and 1750 ppm TBA were selected based on results from a 18-day vapor inhalation study conducted by the National Toxicology Program (NTP, 1997Go). The GC was calibrated by preparing sample bags of known TBA concentrations.

Study design to evaluate TBA-induced {alpha} 2u-nephropathy.
A total of 80 rats (40 males and 40 females) was used in the experiment to assess {alpha}2u-globulin nephropathy ({alpha}2u-N). Male and female rats were assigned to one of two subgroups (five rats/sex/exposure concentration/subgroup) for determination of either histologic or biochemical end points and exposed to target concentrations of 0, 250, 450, or 1750 ppm TBA for 6 h/day for 10 consecutive days.

Tissue preparation for histopathology end points.
At 3.5 days prior to euthanasia, five male and five female rats from each exposure group were subcutaneously implanted with an osmotic pump (Alzet model 2ML1, Alza Corporation, Palo Alto, CA) for delivery of 5-bromo-2-deoxyuridine (BrdU). One day following the final exposure, rats were anesthetized with sodium pentobarbital, and the kidneys were perfused with 2% paraformaldehyde/1% glutaraldehyde via a retrograde intravascular perfusion technique through the descending aorta, as previously described (Larson et al., 1993Go). A midlongitudinal section of the left kidney, midtransverse section of the right kidney, and transverse sections of the duodenum were fixed in a 2% solution of paraformaldehyde and 1% glutaraldehyde. The fixative was replaced after 24 h with 70% ethanol. Tissues were embedded in paraffin and serial sections cut at 5 µm. Tissue sections were stained with H&E and Mallory's Heidenhain stain (Cason, 1950Go) to evaluate histologic lesions and protein droplet accumulation, respectively. Histologic end points evaluated were renal lesions, protein droplet accumulation, quantification of the labeling index (LI) by immunohistochemical staining for BrdU, and determination of {alpha}2u accumulation within renal tubules by {alpha}2u immunohistochemical staining.

Histologic evaluation.
Slides stained with Mallory's Heidenhain, a nonspecific protein stain (Cason, 1950Go), were evaluated according to the relative accumulation or severity of the protein droplets. Protein droplet accumulation was graded on a relative scale of 0–4, based on the size of protein droplets and intensity of staining, with no prior knowledge of treatment group. Scores were assigned to kidney sections from each animal as follows: 0, no protein droplets observed; 1, occasional small protein droplets observed; 2, small protein droplets frequently observed; 3, small to moderately sized protein droplets frequently observed; and 4, large protein droplets seen within many proximal tubular epithelial cells. Lesions were also graded on a scale of 0–4 with no knowledge of treatment group according to the percentage of affected renal tubules throughout the cortex and outer stripe of the outer medulla (OSOM). Scores were assigned to kidney sections from each animal as follows: 0, absence of lesions; 1, < 10% of the tubules affected; 2, 11–25% of the cortex and OSOM involved; 3, 26–50% of the tubules affected; and 4, > 50% of the cortex and OSOM involved. The two individual scores were multiplied to yield a grading scale of 0–16 (Lehman-McKeeman and Caudill, 1999Go).

Cell proliferation.
Immunohistochemical staining for BrdU incorporation was performed on male and female rat kidney and duodenum using a streptavidin alkaline phosphatase technique and Stable Fast Red chromagen. A mouse monoclonal antibody raised toward purified BrdU (Caltag, South San Francisco, CA) and biotin conjugated antimouse IgG (Vector Laboratories, Inc., Burlingame, CA) were used as the primary and secondary antibodies, respectively. A section of duodenum, which normally exhibits a high cell proliferation rate, was included to confirm systematic delivery of BrdU. Tissue sections were processed for BrdU immunohistochemistry according to a modified method of Eldridge et al. (1990) described by Prescott-Mathews et al. (1999). Cell proliferative responses were evaluated through determination of the LI, as described by Prescott-Mathews et al. (1999). The LI was defined as the percentage of positively stained epithelial cells within the proximal tubule. Labeled cells throughout the renal cortex were counted. A total of 2000 tubular epithelial cells were counted within the renal cortex at 400x magnification. In the left kidney, 1000 cells were counted by beginning at the outermost portion of the cortex and counting diagonally inward toward the medulla. Quantification of 1000 cells in the right kidney was accomplished by counting diagonally from the innermost portion of the renal cortex and counting outward.

{alpha} 2u immunohistochemical staining.
Immunohistochemical staining of {alpha}2u was performed on male and female rat kidney sections using a streptavidin horseradish peroxidase technique and aminoethyl carbazole chromagen. A mouse monoclonal antibody (prepared by Hazelton Biotechnologies Co., Vienna, VA) raised toward purified rat urinary {alpha}2u (Kurtz et al., 1976Go) was used as the primary antibody. The mouse monoclonal antibody did not demonstrate any crossreactivity with other proteins in kidney cytosol using SDS-PAGE and Western blot analysis (unpublished observation). {alpha}2u immunohistochemical staining was performed as previously described by Burnett et al. (1989), with modifications described by Prescott-Mathews et al. (1999).

Tissue preparation for biochemical end points.
Five male and five female rats from each exposure group were euthanized by CO2 asphyxiation. Animals were exsanguinated via the caudal vena cava. Kidneys were excised with the capsules removed, weighed, and homogenized in ice-cold 3x (w/v) 0.1 M Na/K phosphate buffer, pH 7.4. Kidney homogenate was stored at –70°C. Prior to measurement of {alpha}2u concentration by the enzyme-linked immunosorbent assay (ELISA), the kidney homogenate was thawed on ice, and kidney cytosol was prepared by centrifugation at 600 x g for 15 min at 4°C. The resultant supernatant was then centrifuged at 116,000 x g for 60 min at 4°C. Aliquots of the supernatant were then stored at –70°C.

Enzyme-linked immunosorbent assay.
The renal concentration of {alpha}2u was measured in male and female rat kidney cytosol using an ELISA with a mouse monoclonal antibody directed against purified rat urinary {alpha}2u (prepared by Hazelton Biotechnologies Co., Vienna, VA). Kidney cytosol prepared from male rats treated for 4 days with 500 mg/kg 2,2,4-trimethylpentane (TMP) (Prescott-Mathews et al., 1997) was used as a positive control on each plate of samples analyzed. Urinary {alpha}2u was purified according to a modified method of Kurtz and coworkers (1976). Using this ELISA technique, {alpha}2u was quantified using anti-mouse IgG alkaline phosphatase conjugate and p-nitrophenyl phosphate, according to a previously described method (Borghoff et al., 1992Go). The {alpha}2u concentration in the kidney cytosol was expressed as micrograms {alpha}2u per milligram total protein. Total protein was measured using the BCA protein assay (Pierce, Rockford, IL), with BSA as the standard.

Study design to evaluate the dosimetry of TBA.
The dosimetry of TBA was assessed in male and female rats following single and repeated (8-day) exposures to target concentrations of 250, 450, or 1750 ppm TBA (n = three rats/sex/exposure concentration/time point) for 6 h/day. Rats were killed at various time points following exposure (2, 4, 6, 8, and 16 h) to measure levels of TBA in the liver, kidney, and blood. These animals were housed in the same chambers as the rats evaluated for {alpha}2u-N. The earliest time point of 2 h following exposure was selected based on coordination of removal of animals from chambers, along with the knowledge that TBA is cleared slowly from blood (half-life ~ 5 h). Although elimination within the first 2 h following the cessation of exposure is critical for pharmacokinetic evaluation, the objective of this study was to compare tissue levels of TBA in male and female rats following single and repeated exposure to TBA. Eight days of repeated exposure was selected based on our ability to handle animals designated to evaluate the various end points described above.

Analysis of tissue TBA levels.
Following exposure, rats were exposed to CO2 (~ 1–1.5 min) at various time points, and blood was withdrawn from the vena cava. Three 1-ml samples were added directly to 10-ml headspace vials and sealed immediately. Three 1-g liver samples and each kidney were minced in 2 ml 0.1 M succinic acid buffer, pH 4.5, in 10-ml headspace vials, and the vial was sealed. Tissue samples were heated at 60°C for 30 min prior to analysis. A 1-ml aliquot of the headspace from each tissue sample was analyzed for TBA using a Headspace Autosampler Model 7694 (Hewlett Packard, Avondale, PA) linked to a Hewlett Packard 5890 Series II gas chromatograph fitted with a capillary injection port, a flame ionization detector, and a Hewlett Packard 3396 Series Integrator. TBA was separated using a series of columns consisting of a capillary SPB-1 precolumn (11 x 0.53 mm; Supelco, Bellefonte Park, PA) connected to a capillary DB-Wax column (30 x 0.53 mm; Alltech, Deerfield, MA) attached to a capillary SPB-5 column (30 x 0.53 mm; Supelco, Bellefonte Park, PA). The oven temperature was initially held at 45°C for 9 min, increased at 70°C/min to 175°C, and held for 4 min. The injection and flame ionization detector temperatures were 200°C and 250°C, respectively. Helium was used as a carrier gas at a flow rate of approximately 4 ml/min at a pressure of 7.3 psi. Individual tissue standard curves were generated by incubating known amounts of TBA with tissue samples obtained from untreated rats under the identical conditions to the unknown samples. The TBA tissue:blood ratio was calculated by dividing the level of TBA in the tissue (liver or kidney) by the level in blood of each rat.

Statistical analysis.
Body and tissue weight comparisons were made using Student's t-test and Tukey-Kramer one-way analysis of variance (ANOVA) with a level of significance of p < 0.05. LI, ELISA, and protein droplet accumulation data were analyzed by Tukey-Kramer one-way ANOVA with a level of significance of p < 0.05. An arcsine transformation was applied to the LI data to meet criteria for normality and homogeneity for ANOVA. The square root of the protein droplet accumulation score (0–16) was used so that the assumption of normality was met. Dunnett's test (p < 0.05) was also carried out to assess the accumulation of protein droplets with exposure concentration.

To analyze trends in protein droplet accumulation and LI with exposure concentration and the correlation between {alpha}2u levels and LI, linear regression analysis was used. Trends were found to be significant, with a level of p < 0.05.

Tissue:blood ratios were compared using one-way ANOVA with a level of significance of p < 0.05.


    RESULTS
 TOP
 ABSTRACT
 INTRODUCTION
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
Chamber Concentrations of TBA
The average daily TBA exposure concentrations (ppm) for a single 6-h exposure, 8 days of repeated 6-h exposures, and 10 days of repeated 6-h exposures are listed in Table 1Go. All but one exposure was reported to be within 10% of the target concentration. The single female rat exposed to 1750 ppm TBA had a higher average and a large standard deviation, which were attributed to electrical power problems on this day caused by a malfunction of the GC sampling system. This caused a transient increase in the TBA flow to the chamber.


View this table:
[in this window]
[in a new window]
 
TABLE 1 Summary of Chamber tert-Butyl Alcohol Concentrations Following a Single Exposure, an 8-Day Exposure, and a 10-Day Exposure to Male and Female F-344 Rats
 
Terminal Body and Organ Weights
Body, liver, and kidney weights were evaluated only in the rats designated for biochemical analysis, as the rats designated for histological analysis had their kidneys perfused. No significant concentration-related differences were observed in terminal body weights in male or female rats (Table 2Go). A statistically significant decrease in the absolute and relative (expressed as a percentage of body weight) liver weight was observed in male rats exposed to 1750 ppm TBA compared to control (p < 0.05), with no consistent exposure-related change in the liver of female rats (Table 2Go). No significant concentration-related difference was observed in absolute kidney weights in male or female rats. Relative kidney weights (expressed as a percentage of body weight) increased in male rats exposed to 1750 ppm and in female rats exposed to 450 and 1750 ppm TBA compared to controls (p < 0.05) (Table 2Go).


View this table:
[in this window]
[in a new window]
 
TABLE 2 Terminal Body and Tissue Weights of Rats Following a 10-Day Exposure to tert-Butyl Alcohol
 
Histopathology
Microscopic lesions in TBA-exposed male rat kidneys were characterized by protein droplet accumulation and karyomegaly within the proximal tubules in male rat kidneys exposed to TBA. Although protein droplets were observed on H&E-stained slides, protein droplets were more readily visualized on kidney sections stained with Mallory's Heidenhain. Minimal protein droplet accumulation (as indicated by the presence of scattered, small, intracytoplasmic droplets) was observed in control male rats; a virtual absence of protein droplets was observed in control or exposed female rats (Fig. 1Go). In male rats exposed to TBA, there was an increased accumulation of protein droplets within the proximal tubule characterized by the formation of large coalescing globules of protein and rare crystalloid protein structures (Fig. 1Go). Grading of Mallory's Heidenhain-stained male rat kidney slides, based on a scale of 0–16, demonstrated an increase in protein droplets at 1750 ppm TBA (Fig. 2Go). Linear regression analysis supported a statistically significant, concentration-dependent, positive trend for the accumulation of protein droplets in male rats exposed to TBA.



View larger version (96K):
[in this window]
[in a new window]
 
FIG. 1. Photomicrographs of Mallory's Heidenhain stained renal cortex from (A) control male rat, (B) TBA-exposed male rat (1750 ppm), and (C) TBA-exposed female rat (1750 ppm). Note the accumulation of large amounts of protein within the proximal tubule and occasional large, coalescing droplet formation (arrow).

 


View larger version (13K):
[in this window]
[in a new window]
 
FIG. 2. Protein droplet accumulation (graded on a scale of 0 to 16 based on severity times the percentage of tubules affected) within the proximal tubule of male rats exposed to TBA. Each symbol represents the score, with the number of rats with that particular score in parentheses. Linear regression analysis supported a statistically significant, concentration-dependent, positive trend (p < 0.05) for protein accumulation. There was also a significant increase in accumulation of protein droplets in the rats exposed to 1750 ppm compared to control rats (p < 0.01).

 
{alpha}2u Immunohistochemistry
{alpha}2u immunohistochemical staining of control and TBA-exposed male rat kidney sections revealed positive staining of protein droplets within renal proximal tubules (Fig. 3Go). No positive staining for {alpha}2u was observed in kidneys from control or TBA-exposed female rats. {alpha}2u-globulin staining was slightly greater in male rats exposed to TBA as compared to control male rats; however, there did not appear to be a TBA exposure-related increase in intensity of {alpha}2u staining in male rats (data not shown).



View larger version (142K):
[in this window]
[in a new window]
 
FIG. 3. Photomicrograph of the renal cortex immunohistochemically stained for {alpha}2u from (A) control male rat and (B) TBA-exposed male rat (1750 ppm).

 
Cell Proliferation
LI were quantified on BrdU immunohistochemically stained kidney sections from male and female rats. When quantification of the LI was restricted to the renal cortex, a concentration-related, statistically significant increase in the LI was observed in all groups of TBA-exposed male rats as compared to control males (Fig. 4Go). No significant differences in LI were observed between TBA-exposed and control female rats.



View larger version (18K):
[in this window]
[in a new window]
 
FIG. 4. Cell proliferation as measured by quantification of the labeling index (LI) in male rats and female rats exposed to TBA for 10 consecutive days. Statistically significant compared to controls; (Tukey-Kramer one-way ANOVA (p < 0.05).

 
Renal Concentration of {alpha}2u
The renal concentration of {alpha}2u was measured in kidney cytosol from male rat kidney using an ELISA. Samples of kidney cytosol from TMP-treated male rats exhibited a statistically significant increase in concentration of {alpha}2u compared to control male rats gavaged with corn oil (Prescott-Mathews et al., 1999Go). These data are shown in the insert to Figure 5Go to demonstrate a positive response from a chemical known to cause {alpha}2u accumulation in the male rat kidney. Kidney cytosol samples from these same male rats treated with TMP served as positive controls in the ELISA assay when samples from TBA-exposed male rats were analyzed. In male rats exposed to 1750 ppm TBA, there was an increase in {alpha}2u concentration as compared to male rats in the control group (p < 0.05; Fig. 5Go). Although the concentration of {alpha}2u in the kidneys of male rats exposed to 250 or 450 ppm TBA was not significantly increased compared to control rats, linear regression analysis revealed a positive correlation (r2 = 0.70) between {alpha}2u concentration and LI in the male rat kidney (Fig. 6Go). The kidneys of female rats have very low levels of {alpha}2u. Because no protein droplets or staining of {alpha}2u was observed in female rats, {alpha}2u concentration was not measured in kidney cytosol of female rats.



View larger version (18K):
[in this window]
[in a new window]
 
FIG. 5. Renal {alpha}2u concentration from male rats exposed to TBA as measured by an ELISA. The inset illustrates the change in {alpha}2u in the kidneys of male rats treated with 500 mg/kg TMP (Prescott-Matthews et al., 1997), which served as a positive control for the ELISA. *Statistically significant compared to control male rats (Tukey-Kramer one-way ANOVA (p < 0.05).

 


View larger version (14K):
[in this window]
[in a new window]
 
FIG. 6. Positive, exposure-related correlation between the labeling index (LI) and renal {alpha}2u concentration for each exposure group. The symbols represent the individual values for each animal at each exposure group (0, 250, 450, and 1750 ppm TBA) (r2 = 0.70).

 
TBA Tissue:Blood Ratios
TBA levels were measured in blood, liver, and kidney following exposure for 1 or 8 days. To compare levels in liver and kidney in male and female rats following 1 and 8 days of exposure, the tissue:blood ratio was calculated. The TBA liver:blood ratio did not change in male or female rats at any exposure concentration or time point of collection when levels from one day were compared to levels following 8 days of repeated exposure (data not shown). However, kidney:blood ratios were increased in male, but not female, rats following 8 consecutive exposure days compared with a single-exposure day (Fig. 7Go). Although the liver:blood ratio remained similar in male and female rats following 8 days of exposure, the kidney:blood ratio was greater in male rats compared with female rats (Fig. 8Go).



View larger version (37K):
[in this window]
[in a new window]
 
FIG. 7. TBA kidney:blood ratio in (A) male and (B) female rats at various times following 1 and 8 days of inhalation exposure to 250, 450, or 1750 ppm TBA. Each bar represents the mean ± SD of three rats.

 


View larger version (34K):
[in this window]
[in a new window]
 
FIG. 8. TBA (A) liver:blood and (B) kidney:blood ratio in male and female rats at various time points following 8 days of inhalation exposure to 250, 450, or 1750 ppm TBA. Each bar represents the mean ± SD of three rats unless otherwise indicated; (a), n = 1; only one rat had a detectable blood concentration of TBA; (b), n = 2; only two rats had detectable levels in blood and kidney samples. *Indicates statistically significant decrease compared to ratio in male rat at the same time (p < 0.05).

 

    DISCUSSION
 TOP
 ABSTRACT
 INTRODUCTION
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
{alpha}2u-N, a syndrome unique to male rats, is characterized by the accumulation of {alpha}2u-containing protein droplets, epithelial cell necrosis, and regenerative cell proliferation within the proximal tubule (U.S. EPA, 1991bGo). The objective of the present study was to determine whether TBA induces {alpha}2u-N and enhanced renal cell proliferation in male, but not female, F-344 rats. When male rats were exposed to TBA for 10 consecutive days, there was an accumulation of protein droplets in the kidney that stained positive for {alpha}2u, and a significant increase in the renal concentration of {alpha}2u. The results from this study show a positive, concentration-dependent correlation between an increase in {alpha}2u and renal cell proliferation that was specific for male rats. However, it should be noted that there was not a significant increase in {alpha}2u at low TBA exposure concentrations, which resulted in an increase in renal cell proliferation. These male rat-specific TBA-induced responses are mild compared to known inducers of {alpha}2u-N such as TMP and d-limonene (Swenberg and Lehman-McKeeman, 1999Go).

Although {alpha}2u immunoreactivity was present in protein droplets in kidneys from TBA-exposed male rats, a linear, concentration-related increase in {alpha}2u staining was not observed. Similar findings were observed in the MTBE 10-day and 13-week inhalation studies in which grading of {alpha}2u-stained kidney sections did not reveal any linear concentration-related increase in {alpha}2u staining in MTBE-exposed male rats (Prescott-Mathews et al., 1999; J. A. Swenberg and D. R. Dietrich, personal communication). In the 10-day study, Prescott-Mathews et al. (1999) observed that the concentration of {alpha}2u increased in kidney cytosol using an ELISA. The same assay was used to show an increase in {alpha}2u concentration in the kidneys of male rats exposed to 1750 ppm TBA for 10 days. A strong {alpha}2u inducer such as TMP caused a greater increase in {alpha}2u accumulation than what was shown for either MTBE or TBA when measured by the ELISA (Prescott-Mathews et al., 1999Go). Staining of protein droplets for {alpha}2u and the level of protein droplet accumulation were also more intense with this chemical (Short et al., 1989Go). The mild increase in {alpha}2u concentration in TBA-exposed male rats may only be detectable by quantitating the level of {alpha}2u using an ELISA, and not by evaluating immunohistochemical staining for {alpha}2u, as the former is a more sensitive method.

Characteristic renal lesions consistent with {alpha}2u-N, such as accumulation of granular casts and linear mineralization of the kidney, are typically observed in studies with continued exposure for 28–90 days. Data from a 90-day drinking water study confirm TBA-induced protein droplet accumulation within the proximal tubule (Lindamood et al., 1992Go). Effects included increased severity of lesions characteristic of chronic progressive nephropathy (greater in female rats) and increased incidence and severity of mineralization (greater in male rats). Both nephropathy and mineralization were reduced at the highest dose level of 4% TBA (3588.5 mg/kg/day) (Table 3Go) compared with the 2% TBA level (1598.9 mg/kg/day), which may have been influenced by the high mortality at 4% level. Protein droplet exacerbation was increased in male rats up to 2% TBA, along with an increase in PCNA-stained S-phase nuclei. Both responses decreased in the rats exposed to 4% TBA in their water, compared with the group exposed to 2% TBA.


View this table:
[in this window]
[in a new window]
 
TABLE 3 Dose Estimates for tert-Butyl Alcohol Studies
 
The NTP (1997) also conducted an 18-day and a 13-week TBA inhalation study in rats. No gross or microscopic changes were observed in rats exposed to 3500 ppm TBA for 18 days (a total of 12 exposure days). Changes in rats exposed to lower concentrations were not evaluated. In the 13-week study, high exposure concentrations to TBA enhanced the severity of chronic nephropathy in male rats. This lesion is a common spontaneous lesion in male rat that is frequently exacerbated by chemical treatment. Exposure of rats to 1080 or 2200 ppm TBA for 13 weeks did not cause exacerbation of protein droplet nephropathy in male rats compared to controls. Review of the NTP archival H&E-stained male rat kidney sections from the 18-day TBA inhalation study revealed an increase in protein droplet accumulation in the proximal tubules of male rats exposed to 450, 900, and 1750 ppm compared to controls, but not to 3500 ppm TBA (unpublished observation). This information was used in designing this 10-day study and selecting exposure concentrations. As only the high exposure concentration (3500 ppm) was originally evaluated in the 18-day study, the ability of TBA to induce protein droplet nephropathy could have been detected with a complete evaluation of all of the exposure groups. In the TBA drinking water study conducted by the NTP, a decrease in protein droplet accumulation was also observed when rats were exposed to high concentrations of TBA (Lindamood et al., 1992Go). Interestingly, cell proliferation, assessed by PCNA staining, followed the change in protein droplet accumulation, with an increase and then a decrease back to control levels at the highest exposure concentration. The decrease in protein droplet accumulation with high exposure concentrations of TBA maybe related to a chemical-specific decrease in protein synthesis or downregulation of {alpha}2u, as is the case following exposure to specific peroxisome proliferators (Corton et al., 1998Go).

The current hypothesis for {alpha}2u-mediated renal tumorigenesis relies on the accumulation of a chemical–{alpha}2u complex resistant to proteolytic degradation in the phagolysosome. As such, {alpha}2u accumulates in and appears to overwhelm the phagolysosome, resulting in cell death and regenerative cell proliferation (Borghoff et al., 1990Go). Enhanced renal cell proliferation, as determined by the LI, was observed in male rats exposed to TBA. Whether {alpha}2u accumulation was totally responsible for the increase in cell proliferation observed only in male rat kidney following exposure to TBA is not clear from this investigation. In Table 3Go, doses were estimated from the various TBA exposure studies and responses compared. In the 90-day drinking water study and this 10-day inhalation study, similar doses (albeit via different routes of administration) apparently induce protein droplet accumulation. In the 2-year chronic study, however, the dose of TBA that caused renal tumors was lower than the dose necessary to cause protein droplets, which may suggest a possible second mechanism of toxicity involved in the tumor formation.

Even though the increase in {alpha}2u concentration correlated with an increase in cell proliferation in this study, when examining the response at the lowest exposure concentration (250 ppm), there was no significant increase in the level of {alpha}2u or the accumulation of protein droplets, whereas the LI was significantly increased. It is possible that measuring the LI in the kidney using a 3-day BrdU pump is a more sensitive method than evaluating protein accumulation or measuring an increase in the concentration of {alpha}2u in the kidney at one time point following exposure. With strong inducers, there is information to demonstrate that the maximum difference in {alpha}2u concentration between controls and treated rats is approximately 24 h following chemical treatment. However, the time course for an increase in {alpha}2u concentration in the kidney following mild inducers may be different. The difference in sensitivity of the assays used for measuring {alpha}2u and the LI, the time course of these responses, or the potential of a second mechanism of toxicity are possible explanations for the lack of correlation between {alpha}2u and the LI at low TBA exposure concentrations.

TBA-induced {alpha}2u-N may be necessary in the development of renal tumors in male rats, but not sufficient. Other factors in addition to {alpha}2u-N that may contribute to the renal cell proliferative response in male rat kidneys following TBA exposure were not obvious from this study. TBA is metabolized to 2-hydroxyisobutyrate and 1,2-methyl-2-propanediol. These metabolites are eliminated in the urine of rats and humans exposed to MTBE or TBA (Amberg et al., 1999Go; Bernauer et al., 1998Go; Miller et al., 1997Go). The presence of these compounds in urine provides evidence that TBA is oxidized. In this investigation, we have shown that TBA appears to accumulate in the male rat kidney with repeated TBA exposure. TBA was also cleared more slowly from the male rat kidney compared with the female rat kidney. Levels of TBA were detected in the male rat kidney 16 h following exposure. Binding of TBA to {alpha}2u would explain this male rat–specific retention of TBA. It is also important to note that the male and female rat kidney were exposed to similar concentrations of TBA; however, an increase in cell proliferation was only observed in the male rat kidney (Fig. 9Go). This demonstrates that TBA itself is not directly responsible for the enhanced cell proliferation that occurs in the male rat kidney.



View larger version (18K):
[in this window]
[in a new window]
 
FIG. 9. Mean labeling index (percentage of control) of male and female rats exposed to 0, 250, 450, or 1750 TBA 6 h/day for 10 days vs. mean kidney TBA levels in rats 2 h following exposure to the same levels of TBA for 8 days. Although these comparisons are not in the same rats exposed to TBA for the same number of days, this relationship demonstrates that the kidneys in female rats achieved levels of TBA that were similar to or greater than the levels in male rat kidneys and show no proliferation.

 
In summary, the present study demonstrates that TBA induces {alpha}2u-N and renal cell proliferation in male, but not female, F-344 rats. Compared to stronger inducers of {alpha}2u-N previously studied, a 10-day exposure to TBA resulted in a mild induction of this syndrome. Although other mechanisms of renal tumor formation have not been definitively ruled out, there are no data to suggest that a mechanism other than {alpha}2u-mediated nephropathy is involved in the low incidence of TBA-induced renal tumors following chronic exposure. Additionally, no increase in {alpha}2u concentration, renal cell proliferation, or renal tumor formation has been demonstrated in female rats exposed to TBA.


    ACKNOWLEDGMENTS
 
The authors gratefully acknowledge Paul Ross and the Animal Care Staff for excellent animal care and maintenance, as well as Kay Roberts, John Murphy, Carl U. Parkinson, Otis Lyght, and Delorise Williams for quality technical assistance. We also thank Drs. Byron Butterworth, Tracy Williams, Leslie Recio, Gregory Kedderis, and Barbara Kuyper for manuscript review. This work was supported by the Oxygenated Fuels Association.


    NOTES
 
1 To whom correspondence should be addressed. Fax: (919) 558-1300. E-mail: borghoff{at}ciit.org. Back


    REFERENCES
 TOP
 ABSTRACT
 INTRODUCTION
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
Amberg, A., Rosner, E., and Dekant, W. (1999). Biotransformation and kinetics of excretion of methyl-tert-butyl ether in rats and humans. Toxicol. Sci. 51, 1–8.[Abstract]

Bernauer, U., Amberg, A., Scheutzow, D., and Dekant, W. (1998). Biotransformation of 12C and 2-13C-labeled methyl tert-butyl ether, ethyl tert-butyl ether, and tert-butyl alcohol in rats: Identification of metabolites in urine by 13C-nuclear magnetic resonance and gas chromatography/mass spectrometry. Chem. Res. Toxicol. 11, 651–658.[ISI][Medline]

Bird, M. G., Burleigh-Flayer, H. D., Chun, J. S., Douglas, J. F., Kneiss, J. J., and Andrews, L. S. (1997). Oncogenicity studies of inhaled methyl tertiary-butyl ether (MTBE) in CD-1 mice and F-344 rats. J. Appl. Toxicol. 17, S45–S55.[ISI][Medline]

Borghoff, S. J., and Lagarde, W. H. (1993). Assessment of binding of 2,4,4-trimethyl-2-pentanol to low-molecular-weight proteins isolated from kidneys of male rats and humans. Toxicol. Appl. Pharmacol. 119, 228–235.[ISI][Medline]

Borghoff, S. J., Short, B. G., and Swenberg, J. A. (1990). Biochemical mechanisms and pathobiology of {alpha}2u-globulin nephropathy. Annu. Rev. Pharmacol. Toxicol. 30, 349–367.[ISI][Medline]

Borghoff, S. J., Youtsey, N. L., and Swenberg, J. A. (1992). A comparison of European high test gasoline and PS-6 unleaded gasoline in their abilities to induce {alpha}2u-globulin nephropathy and renal cell proliferation. Toxicol. Lett. 63, 21–33.[ISI][Medline]

Burnett, V. L., Short, B. G., and Swenberg, J. A. (1989). Localization of {alpha}2u within protein droplets of the male rat kidney: Immunohistochemistry using perfusion-fixed, GMA-embedded tissue sections. J. Histochem. Cytochem. 37, 813–818.[Abstract]

Butterworth B. E., Popp J. A., Conolly R. B., and Goldsworthy, T. L. (1992). Chemically induced cell proliferation in carcinogenesis. In: Mechanisms of Carcinogenesis in Risk Identification (H. Vainio, P N. Magee, D. B., McGregor, and A. J. McMichael, Eds.), pp. 279–305. International Agency for Research on Cancer (IARC), Lyon.

Cason, J. E. (1950). A rapid one-step Mallory-Heidenhain stain for connective tissue. Stain Technol. 25, 225–226.[ISI][Medline]

Cirvello, J. D., Radovsky, A., Heath, J. E., Farnell, D. R., and Lindamood, C. (1995). Toxicity and carcinogenicity of t-butyl alcohol in rats and mice following chronic exposure in drinking water. Toxicol. Ind. Health 11, 151–165.[ISI][Medline]

Corton, J. C., Fan, L.-Q., Brown, S., Anderson, S. P., Bocos, C., Cattley, R. C., Mode, A., and Gustafsson, J. (1998). Down-regulation of cytochrome P450 2C family members and positive acute-phase response gene expression by peroxisome proliferator chemicals. Mol. Pharmacol. 54, 463–473.[Abstract/Free Full Text]

Cosmetic Ingredient Review Expert Panel (CIREP) (1989). Final report on the safety assessment of t-butyl alcohol. J. Am. Coll. Toxicol. 8, 627–641.

Eldridge, S. R., Tilbury, L. F., Goldsworthy, T. L., and Butterworth, B. E. (1990). Measurement of chemically induced cell proliferation in rodent liver and kidney: a comparison of 5-bromo-2'-deoxyuridine and [3H]thymidine administered by injection or osmotic pump. Carcinogenesis 11, 2245–2251.[Abstract]

Kurtz, D. T., Sippel, A. E., and Fiegelson, P. (1976). Effect of thyroid hormones on the level of the hepatic mRNA for {alpha}2u-globulin. Biochemistry 15, 1031–1036.[ISI][Medline]

Larson, J. L., Wolf, D. C., and Butterworth, B. E. (1993). Acute hepatotoxic and nephrotoxic effects of chloroform in male F-344 rats and female B6C3F1 mice. Fundam. Appl. Toxicol. 20, 302–315.[ISI][Medline]

Lehman-McKeeman, L. D., and Caudill, D. (1992). {alpha}2u is the only member of the lipocalin protein superfamily that binds to hyaline droplet inducing agents. Toxicol. Appl. Pharmacol. 116, 170–176.[ISI][Medline]

Lehman-McKeeman, L. D., and Caudill, D. (1999). Development of an in vitro competitive binding assay to predict {alpha}2u–globulin nephropathy. In vitro Mol. Toxicol. 126, 83–95.

Lindamood, C., Farnell, D. R., Giles, H. D., Prejean, J. D., Collins, J. J., Takahashi, K., and Maronpot, R. R. (1992). Subchronic toxicity studies of t-butyl alcohol in rats and mice. Fundam. Appl. Toxicol. 19, 91–100.[ISI][Medline]

McGregor, D. B., Brown, A., Cattanach, P., Edwards, I., McBride, D. and Caspary, W. J. (1988). Responses of the L5178Y tk+/tk mouse lymphoma cell forward assay. II. 18 coded chemicals. Environ. Mol. Mutagen. 11, 91–118.[ISI][Medline]

Miller, M. J., Ferdinandi, E. S., Klan, M., Andrews, L. S., Douglas, J. F., and Kneiss, J. J. (1997). Pharmacokinetics and disposition of methyl t-butyl ether in Fischer-344 rats. J. Appl. Toxicol. 17, S3–12.[ISI][Medline]

Neuhaus, O. W., Flory, W., Biswas, N., and Hollerman, C. E. (1981). Urinary excretion of {alpha}2u-globulin and albumin by adult male rats following treatment with nephrotoxic agents. Nephron 28, 133–140.[ISI][Medline]

NIH (1985). Guide for the care and use of laboratory animals. National Institutes of Health, Publication no. 86-23. U.S. Dept. of Health and Human Services, Public Health Service, Bethesda, MD.

Nihlen, A., Lof, A., and Johanson, G. (1998). Experimental exposure to methyl tertiary-butyl ether. I. Toxicokinetics in humans. Toxicol. Appl. Pharmacol. 148, 274–280.[ISI][Medline]

NTP (1995). Toxicity and carcinogenesis studies of t-butyl alcohol in F-344/N rats and B6C3F1 mice (drinking water study). National Toxicology Program, NTP TR 436. U.S. Dept. of Health and Human Services, Public Health Service, National Institutes of Health.

NTP (1997). Toxicity studies of t-butyl alcohol administered by inhalation to F344/n rats and B6C3F1 mice. National Toxicology Program, NTP TR 53. U.S. Dept. of Health and Human Services, Public Health Service, National Institutes of Health.

Olson, M. J., Johnson, J. T., and Reidy, C. A. (1990). A comparison of male rat and human urinary proteins: Implications for human resistance to hyaline droplet nephropathy. Toxicol. Appl. Pharmacol. 102, 524–536.[ISI][Medline]

Prescott-Mathews, J. S., Poet, T. S., and Borghoff, S. J. (1999). Evaluation of the in vivo interaction of methyl tert-butyl ether with {alpha}2u-globulin in male F-344 rats. Toxicol. Appl. Pharmacol. 157, 60–67.[ISI][Medline]

Short, B. G., Burnett, V. L., and Swenberg, J. A. (1989). Elevated proliferation of proximal tubule cells and localization of accumulated {alpha}2u-globulin in F344 rats during chronic exposure to unleaded gasoline or 2,2,4-trimethylpentane. Toxicol. Appl. Pharmacol. 101, 414–431.[ISI][Medline]

Swenberg, J. A., and Lehman-McKeeman, L. K. (1999). {alpha}2u-Urinary globulin-associated nephropathy as a mechanism of renal tubule cell carcinogenesis in male rats. In Species Differences in Thyroid, Kidney and Urinary Bladder Carcinogenesis (C. C. Capen, E. Dybing, J. M. Rice, and J. D. Wilbourn, Eds.). IARC Scientific Publications No. 147. International Agency for Research on Cancer, Lyon.

Takahashi, K., Lindamood, C., and Maronpot, R. R. (1993). Retrospective study of possible {alpha}-2u-globulin nephropathy and associated cell proliferation in male Fischer 344 rats dosed with t-butyl alcohol. Environ. Heath Perspect. 101 (Suppl. 5), 281–285.[ISI][Medline]

U.S. EPA (1991a). Regulation of fuels and fuel additives. U.S. Environmental Protection Agency. Fed. Register 56, 5352.

U.S. EPA (1991b).{alpha}2u-globulin: association with chemically induced renal toxicity and neoplasia in the male rat. EPA/625/3-019F, U.S. Environmental Protection Agency, Washington, DC.

Williams-Hill, D., Spears, C. P., Prakash, S., Olah, G. A., Shamma, T., Moin, T., Kim, L. Y., and Hill, C. K. (1999). Mutagenicity studies of methyl-tert-butyl ether using the Ames tester strain TA102. Mutat. Res. 446, 15–21.[ISI][Medline]

Zeiger, E., Anderson, B., Haworth, S., Lawlor, T., Mortelmans, K., and Speck, W. (1987). Salmonella mutagenicity tests. III. Results from the testing of 255 chemicals. Environ. Mutagen. 9(Suppl. 9), 1–109.[ISI]