Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Toxicol Pathol ; 51(1-2): 4-14, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36987989

RESUMEN

Ammonium 2,3,3,3-tetrafluoro-2-(heptafluoropropoxy)-propanoate (HFPO-DA) is a short chain member of per- and polyfluoroalkyl substances (PFAS). To better understand the relevance of histopathological effects seen in livers of mice exposed to HFPO-DA for human health risk assessment, histopathological effects were summarized from hematoxylin and eosin (H&E)-stained sections in several repeat-dose toxicity studies in mice. Findings across studies revealed histopathological changes consistent with peroxisomal proliferation, whereas two reports of steatosis could not be confirmed in the published figures. In addition, mechanisms of hepatocellular death were assessed in H&E sections as well as with the apoptotic marker cleaved caspase-3 (CCasp3) in newly cut sections from archived liver blocks from select studies. A comparison of serially CCasp3 immunolabeled and H&E-stained sections revealed that mechanisms of hepatocellular death cannot be clearly discerned in H&E-stained liver sections alone as several examples of putatively necrotic cells were positive for CCasp3. Published whole genome transcriptomic data were also reevaluated for enrichment of various forms of hepatocellular death in response to HFPO-DA, which revealed enrichment of apoptosis and autophagy, but not ferroptosis, pyroptosis, or necroptosis. These morphological and molecular findings are consistent with transcriptomic evidence for peroxisome proliferator-activated receptor alpha (PPARα) signaling in HFPO-DA exposed mice.


Asunto(s)
Carcinoma Hepatocelular , Fluorocarburos , Neoplasias Hepáticas , Ratones , Humanos , Animales , Fluorocarburos/toxicidad
2.
Toxicol Pathol ; 48(3): 494-508, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32138627

RESUMEN

GenX is an alternative to environmentally persistent long-chain perfluoroalkyl and polyfluoroalkyl substances. Mice exposed to GenX exhibit liver hypertrophy, elevated peroxisomal enzyme activity, and other apical endpoints consistent with peroxisome proliferators. To investigate the potential role of peroxisome proliferator-activated receptor alpha (PPARα) activation in mice, and other molecular signals potentially related to observed liver changes, RNA sequencing was conducted on paraffin-embedded liver sections from a 90-day subchronic toxicity study of GenX conducted in mice. Differentially expressed genes were identified for each treatment group, and gene set enrichment analysis was conducted using gene sets that represent biological processes and known canonical pathways. Peroxisome signaling and fatty acid metabolism were among the most significantly enriched gene sets in both sexes at 0.5 and 5 mg/kg GenX; no pathways were enriched at 0.1 mg/kg. Gene sets specific to the PPARα subtype were significantly enriched. These findings were phenotypically anchored to histopathological changes in the same tissue blocks: hypertrophy, mitoses, and apoptosis. In vitro PPARα transactivation assays indicated that GenX activates mouse PPARα. These results indicate that the liver changes observed in GenX-treated mice occur via a mode of action (MOA) involving PPARα, an important finding for human health risk assessment as this MOA has limited relevance to humans.


Asunto(s)
Hidrocarburos Fluorados/toxicidad , Hígado/efectos de los fármacos , PPAR alfa/efectos de los fármacos , Propionatos/toxicidad , Animales , Femenino , Humanos , Masculino , Ratones , Medición de Riesgo , Transcriptoma/efectos de los fármacos
3.
J Appl Toxicol ; 39(9): 1267-1282, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31215065

RESUMEN

Ammonium 2,3,3,3-tetrafluoro-2-(heptafluoropropoxy)-propanoate, also known as GenX, is a processing aid used in the manufacture of fluoropolymers. GenX is one of several chemistries developed as an alternative to long-chain poly-fluoroalkyl substances, which tend to have long clearance half-lives and are environmentally persistent. Unlike poly-fluoroalkyl substances, GenX has more rapid clearance, but has been detected in US and international water sources. There are currently no federal drinking water standards for GenX in the USA; therefore, we developed a non-cancer oral reference dose (RfD) for GenX based on available repeated dose studies. The review of the available data indicate that GenX is unlikely to be genotoxic. A combination of traditional frequentist benchmark dose models and Bayesian benchmark dose models were used derive relevant points of departure from mammalian toxicity studies. In addition, deterministic and probabilistic RfD values were developed using available tools and regulatory guidance. The two approaches resulted in a narrow range of RfD values for liver lesions observed in a 2-year bioassay in rats (0.01-0.02 mg/kg/day). The probabilistic approach resulted in the lower, i.e., more conservative RfD. The probabilistic RfD of 0.01 mg/kg/day results in a maximum contaminant level goal of 70 ppb. It is anticipated that these values, along with the hazard identification and dose-response modeling described herein, should be informative for risk assessors and regulators interested in setting health-protective drinking water guideline values for GenX.


Asunto(s)
Benchmarking , Agua Potable/normas , Hidrocarburos Fluorados/toxicidad , Nivel sin Efectos Adversos Observados , Propionatos/toxicidad , Estándares de Referencia , Contaminantes Químicos del Agua/toxicidad , Animales , Humanos , Dosificación Letal Mediana , Modelos Animales , Ratas , Estados Unidos
4.
J Appl Toxicol ; 38(3): 351-365, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29064106

RESUMEN

The current US Environmental Protection Agency (EPA) reference dose (RfD) for oral exposure to chromium, 0.003 mg kg-1  day-1 , is based on a no-observable-adverse-effect-level from a 1958 bioassay of rats exposed to ≤25 ppm hexavalent chromium [Cr(VI)] in drinking water. EPA characterizes the confidence in this RfD as "low." A more recent cancer bioassay indicates that Cr(VI) in drinking water is carcinogenic to mice at ≥30 ppm. To assess whether the existing RfD is health protective, neoplastic and non-neoplastic lesions from the 2 year cancer bioassay were modeled in a three-step process. First, a rodent physiological-based pharmacokinetic (PBPK) model was used to estimate internal dose metrics relevant to each lesion. Second, benchmark dose modeling was conducted on each lesion using the internal dose metrics. Third, a human PBPK model was used to estimate the daily mg kg-1 dose that would produce the same internal dose metric in both normal and susceptible humans. Mechanistic research into the mode of action for Cr(VI)-induced intestinal tumors in mice supports a threshold mechanism involving intestinal wounding and chronic regenerative hyperplasia. As such, an RfD was developed using incidence data for the precursor lesion diffuse epithelial hyperplasia. This RfD was compared to RfDs for other non-cancer endpoints; all RfD values ranged 0.003-0.02 mg kg-1  day-1 . The lowest of these values is identical to EPA's existing RfD value. Although the RfD value remains 0.003 mg kg-1  day-1 , the confidence is greatly improved due to the use of a 2-year bioassay, mechanistic data, PBPK models and benchmark dose modeling.


Asunto(s)
Bioensayo , Pruebas de Carcinogenicidad/métodos , Cromo/toxicidad , Contaminantes Ambientales/toxicidad , Neoplasias Intestinales/inducido químicamente , Modelos Biológicos , Administración Oral , Animales , Bioensayo/normas , Calibración , Pruebas de Carcinogenicidad/normas , Cromo/administración & dosificación , Cromo/farmacocinética , Relación Dosis-Respuesta a Droga , Contaminantes Ambientales/administración & dosificación , Contaminantes Ambientales/farmacocinética , Femenino , Humanos , Neoplasias Intestinales/patología , Masculino , Ratones , Nivel sin Efectos Adversos Observados , Ratas , Estándares de Referencia , Medición de Riesgo , Especificidad de la Especie , Estados Unidos , United States Environmental Protection Agency
5.
Int J Toxicol ; 37(2): 125-143, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29357719

RESUMEN

The National Academy of Science has recommended that a risk of bias (RoB; credibility of the link between exposure and outcome) assessment be conducted on studies that are used as primary data sources for hazard identification and dose-response assessment. Few applications of such have been conducted. Using trichloroethylene and congenital heart defects (CHDs) as a case study, we explore the role of RoB in chemical risk assessment using the National Toxicology Program's Office of Health Assessment and Translation RoB tool. Selected questions were tailored to evaluation of CHD and then applied to 12 experimental animal studies and 9 epidemiological studies. Results demonstrated that the inconsistent findings of a single animal study were likely explained by the limitations in study design assessed via RoB (eg, lack of concurrent controls, unvalidated method for assessing outcome, unreliable statistical methods, etc). Such limitations considered in the context of the body of evidence render the study not sufficiently reliable for the development of toxicity reference values. The case study highlights the utility of RoB as part of a robust risk assessment process and specifically demonstrates the role RoB can play in objectively selecting candidate data sets to develop toxicity values.


Asunto(s)
Cardiopatías Congénitas , Revisiones Sistemáticas como Asunto , Tricloroetileno , Animales , Femenino , Humanos , Embarazo , Sesgo , Cardiopatías Congénitas/inducido químicamente , Cardiopatías Congénitas/epidemiología , Intercambio Materno-Fetal , Medición de Riesgo/métodos , Solventes/toxicidad , Tricloroetileno/toxicidad
6.
Toxicol Pathol ; 45(8): 1091-1101, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29161989

RESUMEN

High concentrations of hexavalent chromium (Cr(VI)), captan, and folpet induce duodenal tumors in mice. Using standardized tissue collection procedures and diagnostic criteria, we compared the duodenal histopathology in B6C3F1 mice following exposure to these 3 carcinogens to determine whether they share similar histopathological characteristics. B6C3F1 mice ( n = 20 per group) were exposed to 180 ppm Cr(VI) in drinking water, 12,000 ppm captan in feed, or 16,000 ppm folpet in feed for 28 days. After 28 days of exposure, villous enterocyte hypertrophy and mild crypt epithelial hyperplasia were observed in all exposed mice. In a subset of mice allowed to recover for 28 days, duodenal samples were generally indistinguishable from those of unexposed mice. Changes in the villi and lack of observable damage to the crypt compartment suggest that toxicity was mediated in the villi, which is consistent with earlier studies on each chemical. These findings indicate that structurally diverse agents can induce similar (and reversible) phenotypic changes in the duodenum. These intestinal carcinogens likely converge on common pathways involving irritation and wounding of the villi leading to crypt regenerative hyperplasia that, under protracted high-dose exposure scenarios, increases the risk of spontaneous mutation and tumorigenesis.


Asunto(s)
Captano/toxicidad , Carcinógenos/toxicidad , Cromo/toxicidad , Duodeno/efectos de los fármacos , Duodeno/patología , Ftalimidas/toxicidad , Administración Oral , Animales , Relación Dosis-Respuesta a Droga , Femenino , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Ratones Endogámicos
7.
Toxicol Appl Pharmacol ; 306: 120-33, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27396814

RESUMEN

To extend previous models of hexavalent chromium [Cr(VI)] reduction by gastric fluid (GF), ex vivo experiments were conducted to address data gaps and limitations identified with respect to (1) GF dilution in the model; (2) reduction of Cr(VI) in fed human GF samples; (3) the number of Cr(VI) reduction pools present in human GF under fed, fasted, and proton pump inhibitor (PPI)-use conditions; and (4) an appropriate form for the pH-dependence of Cr(VI) reduction rate constants. Rates and capacities of Cr(VI) reduction were characterized in gastric contents from fed and fasted volunteers, and from fasted pre-operative patients treated with PPIs. Reduction capacities were first estimated over a 4-h reduction period. Once reduction capacity was established, a dual-spike approach was used in speciated isotope dilution mass spectrometry analyses to characterize the concentration-dependence of the 2nd order reduction rate constants. These data, when combined with previously collected data, were well described by a three-pool model (pool 1 = fast reaction with low capacity; pool 2 = slow reaction with higher capacity; pool 3 = very slow reaction with higher capacity) using pH-dependent rate constants characterized by a piecewise, log-linear relationship. These data indicate that human gastric samples, like those collected from rats and mice, contain multiple pools of reducing agents, and low concentrations of Cr(VI) (<0.7 mg/L) are reduced more rapidly than high concentrations. The data and revised modeling results herein provide improved characterization of Cr(VI) gastric reduction kinetics, critical for Cr(VI) pharmacokinetic modeling and human health risk assessment.


Asunto(s)
Cromo/química , Jugo Gástrico/química , Modelos Biológicos , Contaminantes Químicos del Agua/química , Ayuno , Humanos , Oxidación-Reducción
8.
J Appl Toxicol ; 35(11): 1292-308, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25825072

RESUMEN

Tetrabromobisphenol A (TBBPA) is used in a diverse array of products to improve fire safety. The National Toxicology Program (NTP) recently completed a 2-year bioassay for TBBPA. The objective of the present study was to develop a cancer-based and a non-cancer based toxicity value and to compare such to appropriate estimates of human exposure. Data from the NTP 2-year and 13-week studies were selected to develop candidate toxicity values. Benchmark dose modeling and subsequent evaluation of candidate values resulted in selection of an oral reference dose (RfD) of 0.6 mg kg(-1) day(-1) based on uterine hyperplasia in rats and an oral cancer slope factor (OSF) of 0.00315 per mg kg(-1) day(-1) based on an increased incidence of uterine tumors in rats. Lifetime average daily dose (LADD) estimates ranged from 2.2 E(-7) to 3.9 E(-6) mg kg(-1) day(-1) based on age-adjusted exposures to TBBPA via breast milk consumption, dietary intake, soil/dust ingestion and drinking water ingestion in infants, young children, older children and adults. Average daily dose (ADD) estimates ranged from 3.2 E (-7) to 8.4 E(-5) mg kg(-1) day(-1). Resulting margin of exposure (MOE) values were > 800 000 for non-cancer endpoints and > 32,000,000 for cancer-based endpoints. These data collectively indicate a low level of health concern associated with exposures to TBBPA based on current data. It is anticipated that the exposure estimates, along with the toxicity values described within, should be informative for understanding human health hazards associated with TBBPA.


Asunto(s)
Exposición a Riesgos Ambientales/efectos adversos , Bifenilos Polibrominados/toxicidad , Administración Oral , Adolescente , Adulto , Animales , Niño , Preescolar , Relación Dosis-Respuesta a Droga , Agua Potable/análisis , Polvo/análisis , Determinación de Punto Final , Femenino , Humanos , Hiperplasia/inducido químicamente , Hiperplasia/patología , Lactante , Masculino , Ratones , Leche Humana/química , Ratas , Pruebas de Toxicidad , Neoplasias Uterinas/inducido químicamente , Neoplasias Uterinas/patología
9.
J Appl Toxicol ; 34(5): 525-36, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-23943231

RESUMEN

High concentrations of hexavalent chromium [Cr(VI)] in drinking water induce villous cytotoxicity and compensatory crypt hyperplasia in the small intestines of mice (but not rats). Lifetime exposure to such cytotoxic concentrations increases intestinal neoplasms in mice, suggesting that the mode of action for Cr(VI)-induced intestinal tumors involves chronic wounding and compensatory cell proliferation of the intestine. Therefore, we developed a chronic oral reference dose (RfD) designed to be protective of intestinal damage and thus intestinal cancer. A physiologically based pharmacokinetic model for chromium in mice was used to estimate the amount of Cr(VI) entering each intestinal tissue section (duodenum, jejunum and ileum) from the lumen per day (normalized to intestinal tissue weight). These internal dose metrics, together with corresponding incidences for diffuse hyperplasia, were used to derive points of departure using benchmark dose modeling and constrained nonlinear regression. Both modeling techniques resulted in similar points of departure, which were subsequently converted to human equivalent doses using a human physiologically based pharmacokinetic model. Applying appropriate uncertainty factors, an RfD of 0.006 mg kg(-1) day(-1) was derived for diffuse hyperplasia-an effect that precedes tumor formation. This RfD is protective of both noncancer and cancer effects in the small intestine and corresponds to a safe drinking water equivalent level of 210 µg l(-1). This concentration is higher than the current federal maximum contaminant level for total Cr (100 µg l(-1)) and well above levels of Cr(VI) in US drinking water supplies (typically ≤ 5 µg l(-1)).


Asunto(s)
Pruebas de Carcinogenicidad/métodos , Cromo/toxicidad , Agua Potable/normas , Neoplasias Intestinales/inducido químicamente , Pruebas de Toxicidad Crónica/métodos , Contaminantes Químicos del Agua/toxicidad , Animales , Pruebas de Carcinogenicidad/normas , Relación Dosis-Respuesta a Droga , Agua Potable/química , Femenino , Humanos , Hiperplasia , Neoplasias Intestinales/patología , Masculino , Ratones , Modelos Biológicos , Nivel sin Efectos Adversos Observados , Estándares de Referencia , Factores Sexuales , Pruebas de Toxicidad Crónica/normas , Estados Unidos , United States Environmental Protection Agency
10.
Environ Mol Mutagen ; 65(3-4): 129-136, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38717101

RESUMEN

Chronic exposure to high (20,000 ppm) concentrations of tert-butyl alcohol (TBA) in drinking water, equivalent to ~2100 mg/kg bodyweight per day, is associated with slight increases in the incidence of thyroid follicular cell adenomas and carcinomas in mice, with no other indications of carcinogenicity. In a recent toxicological review of TBA, the U.S. EPA determined that the genotoxic potential of TBA was inconclusive, largely based on non-standard studies such as in vitro comet assays. As such, the potential role of genotoxicity in the mode of action of thyroid tumors and therefore human relevance was considered uncertain. To address the potential role of genotoxicity in TBA-associated thyroid tumor formation, CD-1 mice were exposed up to a maximum tolerated dose of 1500 mg/kg-day via oral gavage for two consecutive days and DNA damage was assessed with the comet assay in the thyroid. Blood TBA levels were analyzed by headspace GC-MS to confirm systemic tissue exposure. At study termination, no significant increases (DNA breakage) or decreases (DNA crosslinks) in %DNA tail were observed in TBA exposed mice. In contrast, oral gavage of the positive control ethyl methanesulfonate significantly increased %DNA tail in the thyroid. These findings are consistent with most genotoxicity studies on TBA and provide mechanistic support for non-linear, threshold toxicity criteria for TBA. While the mode of action for the thyroid tumors remains unclear, linear low dose extrapolation methods for TBA appear more a matter of policy than science.


Asunto(s)
Ensayo Cometa , Daño del ADN , Glándula Tiroides , Alcohol terc-Butílico , Animales , Ensayo Cometa/métodos , Ratones , Alcohol terc-Butílico/toxicidad , Daño del ADN/efectos de los fármacos , Glándula Tiroides/efectos de los fármacos , Glándula Tiroides/patología , Neoplasias de la Tiroides/inducido químicamente , Neoplasias de la Tiroides/patología , Mutágenos/toxicidad , Masculino , Femenino
11.
Toxicol Sci ; 200(1): 165-182, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38574381

RESUMEN

Like many per- or polyfluorinated alkyl substances (PFAS), toxicity studies with HFPO-DA (ammonium, 2,3,3,3-tetrafluoro-2-(heptafluoropropoxy)-propanoate), a short-chain PFAS used in the manufacture of some types of fluorinated polymers, indicate that the liver is the primary target of toxicity in rodents following oral exposure. Although the current weight of evidence supports the PPARα mode of action (MOA) for liver effects in HFPO-DA-exposed mice, alternate MOAs have also been hypothesized including PPARγ or cytotoxicity. To further evaluate the MOA for HFPO-DA in rodent liver, transcriptomic analyses were conducted on samples from primary mouse, rat, and pooled human hepatocytes treated for 12, 24, or 72 h with various concentrations of HFPO-DA, or agonists of PPARα (GW7647), PPARγ (rosiglitazone), or cytotoxic agents (ie, acetaminophen or d-galactosamine). Concordance analyses of enriched pathways across chemicals within each species demonstrated the greatest concordance between HFPO-DA and PPARα agonist GW7647-treated hepatocytes compared with the other chemicals evaluated. These findings were supported by benchmark concentration modeling and predicted upstream regulator results. In addition, transcriptomic analyses across species demonstrated a greater transcriptomic response in rodent hepatocytes treated with HFPO-DA or agonists of PPARα or PPARγ, indicating rodent hepatocytes are more sensitive to HFPO-DA or PPARα/γ agonist treatment. These results are consistent with previously published transcriptomic analyses and further support that liver effects in HFPO-DA-exposed rodents are mediated through rodent-specific PPARα signaling mechanisms as part of the MOA for PPARα activator-induced rodent hepatocarcinogenesis. Thus, effects observed in mouse liver are not appropriate endpoints for toxicity value development for HFPO-DA in human health risk assessment.


Asunto(s)
Hepatocitos , PPAR alfa , PPAR gamma , Transcriptoma , Animales , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , PPAR alfa/agonistas , PPAR alfa/genética , PPAR alfa/metabolismo , Humanos , PPAR gamma/genética , PPAR gamma/agonistas , PPAR gamma/metabolismo , Transcriptoma/efectos de los fármacos , Masculino , Ratones , Fluorocarburos/toxicidad , Ratas , Propionatos/toxicidad , Células Cultivadas , Perfilación de la Expresión Génica , Rosiglitazona/farmacología , Rosiglitazona/toxicidad , Ratas Sprague-Dawley , Ratones Endogámicos C57BL , Especificidad de la Especie , Relación Dosis-Respuesta a Droga , Butiratos , Compuestos de Fenilurea
12.
Toxicol Sci ; 200(1): 183-198, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38574385

RESUMEN

Recent in vitro transcriptomic analyses for the short-chain polyfluoroalkyl substance, HFPO-DA (ammonium, 2,3,3,3-tetrafluoro-2-(heptafluoropropoxy)-propanoate), support conclusions from in vivo data that HFPO-DA-mediated liver effects in mice are part of the early key events of the peroxisome proliferator-activated receptor alpha (PPARα) activator-induced rodent hepatocarcinogenesis mode of action (MOA). Transcriptomic responses in HFPO-DA-treated rodent hepatocytes have high concordance with those treated with a PPARα agonist and lack concordance with those treated with PPARγ agonists or cytotoxic agents. To elucidate whether HFPO-DA-mediated transcriptomic responses in mouse liver are PPARα-dependent, additional transcriptomic analyses were conducted on samples from primary PPARα knockout (KO) and wild-type (WT) mouse hepatocytes exposed for 12, 24, or 72 h with various concentrations of HFPO-DA, or well-established agonists of PPARα (GW7647) and PPARγ (rosiglitazone), or cytotoxic agents (acetaminophen or d-galactosamine). Pathway and predicted upstream regulator-level responses were highly concordant between HFPO-DA and GW7647 in WT hepatocytes. A similar pattern was observed in PPARα KO hepatocytes, albeit with a distinct temporal and concentration-dependent delay potentially mediated by compensatory responses. This delay was not observed in PPARα KO hepatocytes exposed to rosiglitazone, acetaminophen, d-galactosamine. The similarity in transcriptomic signaling between HFPO-DA and GW7647 in both the presence and absence of PPARα in vitro indicates these compounds share a common MOA.


Asunto(s)
Hepatocitos , Ratones Noqueados , PPAR alfa , PPAR gamma , Transcriptoma , Animales , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , PPAR alfa/agonistas , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR gamma/agonistas , PPAR gamma/genética , PPAR gamma/metabolismo , Transcriptoma/efectos de los fármacos , Ratones , Fluorocarburos/toxicidad , Propionatos/farmacología , Propionatos/toxicidad , Ratones Endogámicos C57BL , Masculino , Células Cultivadas , Perfilación de la Expresión Génica , Acetaminofén/toxicidad , Citotoxinas/toxicidad , Butiratos , Compuestos de Fenilurea
13.
Crit Rev Toxicol ; 43(3): 244-74, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23445218

RESUMEN

Abstract Chronic exposure to high concentrations of hexavalent chromium (Cr(VI)) in drinking water causes intestinal adenomas and carcinomas in mice, but not in rats. Cr(VI) causes damage to intestinal villi and crypt hyperplasia in mice after only one week of exposure. After two years of exposure, intestinal damage and crypt hyperplasia are evident in mice (but not rats), as are intestinal tumors. Although Cr(VI) has genotoxic properties, these findings suggest that intestinal tumors in mice arise as a result of chronic mucosal injury. To better understand the mode of action (MOA) of Cr(VI) in the intestine, a 90-day drinking water study was conducted to collect histological, biochemical, toxicogenomic and pharmacokinetic data in intestinal tissues. Using MOA analyses and human relevance frameworks proposed by national and international regulatory agencies, the weight of evidence supports a cytotoxic MOA with the following key events: (a) absorption of Cr(VI) from the intestinal lumen, (b) toxicity to intestinal villi, (c) crypt regenerative hyperplasia and (d) clonal expansion of mutations within the crypt stem cells, resulting in late onset tumorigenesis. This article summarizes the data supporting each key event in the MOA, as well as data that argue against a mutagenic MOA for Cr(VI)-induced intestinal tumors.


Asunto(s)
Cromo/toxicidad , Neoplasias Intestinales/inducido químicamente , Intestino Delgado/efectos de los fármacos , Animales , Cromo/administración & dosificación , Cromo/farmacocinética , Cromo/envenenamiento , Agua Potable , Humanos , Neoplasias Intestinales/metabolismo , Intestino Delgado/metabolismo , Ratones , Ratas
14.
Mutat Res ; 754(1-2): 15-21, 2013 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-23583686

RESUMEN

Chronic exposure to high concentrations of hexavalent chromium [Cr(VI)] as sodium dichromate dihydrate (SDD) in drinking water induces duodenal tumors in mice, but the mode of action (MOA) for these tumors has been a subject of scientific debate. To evaluate the tumor-site-specific genotoxicity and cytotoxicity of SDD in the mouse small intestine, tissue pathology and cytogenetic damage were evaluated in duodenal crypt and villus enterocytes from B6C3F1 mice exposed to 0.3-520mg/L SDD in drinking water for 7 and 90 days. Allele-competitive blocker PCR (ACB-PCR) was used to investigate the induction of a sensitive, tumor-relevant mutation, specifically in vivo K-Ras codon 12 GAT mutation, in scraped duodenal epithelium following 90 days of drinking water exposure. Cytotoxicity was evident in the villus as disruption of cellular arrangement, desquamation, nuclear atypia and blunting. Following 90 days of treatment, aberrant nuclei, occurring primarily at villi tips, were significantly increased at ≥60mg/L SDD. However, in the crypt compartment, there were no dose-related effects on mitotic and apoptotic indices or the formation of aberrant nuclei indicating that Cr(VI)-induced cytotoxicity was limited to the villi. Cr(VI) caused a dose-dependent proliferative response in the duodenal crypt as evidenced by an increase in crypt area and increased number of crypt enterocytes. Spontaneous K-Ras codon 12 GAT mutations in untreated mice were higher than expected, in the range of 10(-2) to 10(-3); however no treatment-related trend in the K-Ras codon 12 GAT mutation was observed. The high spontaneous background K-Ras mutant frequency and Cr(VI) dose-related increases in crypt enterocyte proliferation, without dose-related increase in K-Ras mutant frequency, micronuclei formation, or change in mitotic or apoptotic indices, are consistent with a lack of genotoxicity in the crypt compartment, and a MOA involving accumulation of mutations late in carcinogenesis as a consequence of sustained regenerative proliferation.


Asunto(s)
Cromo/toxicidad , Agua Potable , Duodeno/efectos de los fármacos , Genes ras , Pruebas de Micronúcleos , Mutación , Animales , Secuencia de Bases , Codón , Cartilla de ADN , Duodeno/metabolismo , Femenino , Ratones , Reacción en Cadena de la Polimerasa
15.
Regul Toxicol Pharmacol ; 66(2): 208-16, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23545073

RESUMEN

The need to remediate contaminated soils is typically accomplished by applying standard risk assessment methods followed by risk management to select remedial options. These human health risk assessments (HHRAs) have been largely conducted in a formulaic manner that relies heavily on standard deterministic exposure, toxicity assumptions and fixed mathematical formulas. The HHRA approach, with its traditional formulaic practice, does not take advantage of problem formulation in the same manner as is done in ecological risk assessment, and historically, has generally failed to emphasize incorporation of site-specific information. In response to these challenges, the National Academy of Sciences recently made several recommendations regarding the conduct of HHRAs, one of which was to begin all such assessments with problem formulation. These recommendations have since been extended to dose response assessment. In accordance with these recommendations, a group of experts presented and discussed findings that highlighted the importance and impact of including problem formulation when determining the need for remediation of dioxin contamination in soils, focusing in particular on exposure assessment is described.


Asunto(s)
Carcinógenos/toxicidad , Dioxinas/toxicidad , Exposición a Riesgos Ambientales/efectos adversos , Contaminantes del Suelo/toxicidad , Adulto , Carcinógenos/farmacocinética , Niño , Dioxinas/farmacocinética , Exposición a Riesgos Ambientales/análisis , Humanos , Modelos Teóricos , Medición de Riesgo/métodos , Contaminantes del Suelo/farmacocinética
16.
J Appl Toxicol ; 33(12): 1395-406, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22936336

RESUMEN

Sulfolane is a widely used industrial solvent that is often used for gas treatment (sour gas sweetening; hydrogen sulfide removal from shale and coal processes, etc.), and in the manufacture of polymers and electronics, and may be found in pharmaceuticals as a residual solvent used in the manufacturing processes. Sulfolane is considered a high production volume chemical with worldwide production around 18 000-36 000 tons per year. Given that sulfolane has been detected as a contaminant in groundwater, an important potential route of exposure is tap water ingestion. Because there are currently no federal drinking water standards for sulfolane in the USA, we developed a noncancer oral reference dose (RfD) based on benchmark dose modeling, as well as a tap water screening value that is protective of ingestion. Review of the available literature suggests that sulfolane is not likely to be mutagenic, clastogenic or carcinogenic, or pose reproductive or developmental health risks except perhaps at very high exposure concentrations. RfD values derived using benchmark dose modeling were 0.01-0.04 mg kg(-1) per day, although modeling of developmental endpoints resulted in higher values, approximately 0.4 mg kg(-1) per day. The lowest, most conservative, RfD of 0.01 mg kg(-1) per day was based on reduced white blood cell counts in female rats. This RfD was used to develop a tap water screening level that is protective of ingestion, viz. 365 µg l(-1). It is anticipated that these values, along with the hazard identification and dose-response modeling described herein, should be informative for risk assessors and regulators interested in setting health-protective drinking water guideline values for sulfolane.


Asunto(s)
Benchmarking , Agua Potable , Modelos Teóricos , Tiofenos , Pruebas de Toxicidad , Contaminantes Químicos del Agua , Administración Oral , Animales , Relación Dosis-Respuesta a Droga , Agua Potable/análisis , Agua Potable/normas , Agua Subterránea/química , Concentración Máxima Admisible , Nivel sin Efectos Adversos Observados , Valores de Referencia , Especificidad de la Especie , Tiofenos/análisis , Tiofenos/toxicidad , Contaminantes Químicos del Agua/análisis , Contaminantes Químicos del Agua/toxicidad
17.
Birth Defects Res ; 115(11): 1011-1062, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37219003

RESUMEN

BACKGROUND: Some per- and poly-fluoroalkyl substances (PFAS) cause neonatal mortality and lower birth weight in rodents. We constructed an Adverse Outcome Pathway (AOP) network for neonatal mortality and lower birth weight in rodents, comprising three putative AOPs. We then assessed strengths of the evidence for the AOPs and applicability to PFAS. Finally, we considered the relevance of this AOP network to human health. METHODS: Literature searches targeted PFAS, peroxisome proliferator-activated receptor (PPAR) agonists, other nuclear receptors, relevant tissues, and developmental targets. We used reviews of established biology and described results of studies with prenatal PFAS exposure that assessed birth weight and neonatal survival. Molecular initiating events (MIEs) and key events (KEs) were proposed and strengths of KE relationships (KERs), applicability to PFAS, and human relevance were assessed. RESULTS: Neonatal mortality has been observed in rodents following gestational exposure to most longer chain PFAS studied, often coincident with lower birth weight. In AOP 1, PPARα activation and PPARγ activation or downregulation are MIEs; placental insufficiency, fetal nutrient restriction, neonatal hepatic glycogen deficit, and hypoglycemia are KEs leading to neonatal mortality and lower birth weight. In AOP 2, constitutive androstane receptor (CAR) and pregnane X receptor (PXR) activation upregulates Phase II metabolism, lowering maternal circulating thyroid hormones. In AOP 3, disrupted pulmonary surfactant function and PPARγ downregulation cause neonatal airway collapse and mortality from respiratory failure. CONCLUSIONS: It is likely that different components of this AOP network will apply to different PFAS, largely determined by which nuclear receptors they activate. The MIEs and KEs in this AOP network can occur in humans, but differences in PPAR structure and function, and the timeline of liver and lung development, suggest that humans may be less susceptible to this AOP network. This putative AOP network elucidates knowledge gaps and research needed to better understand the developmental toxicity of PFAS.


Asunto(s)
Fluorocarburos , Roedores , Recién Nacido , Animales , Humanos , Embarazo , Femenino , Peso al Nacer , PPAR gamma , Placenta , Mortalidad Infantil , Fluorocarburos/toxicidad
18.
Toxicol Sci ; 192(1): 15-29, 2023 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-36629480

RESUMEN

HFPO-DA (ammonium, 2,3,3,3-tetrafluoro-2-(heptafluoropropoxy)propanoate) is a short-chain polyfluorinated alkyl substance (PFAS) used in the manufacture of some types of fluorinated polymers. Like many PFAS, toxicity studies with HFPO-DA indicate the liver is the primary target of toxicity in rodents following oral exposure. Due to the structural diversity of PFAS, the mode of action (MOA) can differ between PFAS for the same target tissue. There is significant evidence for involvement of peroxisome proliferator-activated receptor alpha (PPARα) activation based on molecular and histopathological responses in the liver following HFPO-DA exposure, but other MOAs have also been hypothesized based on limited evidence. The MOA underlying the liver effects in mice exposed to HFPO-DA was assessed in the context of the Key Events (KEs) outlined in the MOA framework for PPARα activator-induced rodent hepatocarcinogenesis. The first 3 KEs (ie, PPARα activation, alteration of cell growth pathways, and perturbation of cell growth/survival) are supported by several lines of evidence from both in vitro and in vivo data available for HFPO-DA. In contrast, alternate MOAs, including cytotoxicity, PPARγ and mitochondrial dysfunction are generally not supported by the scientific literature. HFPO-DA-mediated liver effects in mice are not expected in humans as only KE 1, PPARα activation, is shared across species. PPARα-mediated gene expression in humans produces only a subset (ie, lipid modulating effects) of the responses observed in rodents. As such, the adverse effects observed in rodent livers should not be used as the basis of toxicity values for HFPO-DA for purposes of human health risk assessment.


Asunto(s)
Fluorocarburos , Neoplasias Hepáticas , Humanos , Ratones , Animales , PPAR alfa/genética , PPAR alfa/metabolismo , Fluorocarburos/toxicidad , Hígado , Neoplasias Hepáticas/metabolismo , Roedores/metabolismo
19.
Toxicol Appl Pharmacol ; 259(1): 13-26, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22155349

RESUMEN

Chronic administration of high doses of hexavalent chromium [Cr(VI)] as sodium dichromate dihydrate (SDD) elicits alimentary cancers in mice. To further elucidate key events underlying tumor formation, a 90-day drinking water study was conducted in B6C3F1 mice. Differential gene expression was examined in duodenal and jejunal epithelial samples following 7 or 90days of exposure to 0, 0.3, 4, 14, 60, 170 or 520mg/L SDD in drinking water. Genome-wide microarray analyses identified 6562 duodenal and 4448 jejunal unique differentially expressed genes at day 8, and 4630 and 4845 unique changes, respectively, in the duodenum and jejunum at day 91. Comparative analysis identified significant overlap in duodenal and jejunal differential gene expression. Automated dose-response modeling identified >80% of the differentially expressed genes exhibited sigmoidal dose-response curves with EC(50) values ranging from 10 to 100mg/L SDD. Only 16 genes satisfying the dose-dependent differential expression criteria had EC(50) values <10mg/L SDD, 3 of which were regulated by Nrf2, suggesting oxidative stress in response to SDD at low concentrations. Analyses of differentially expressed genes identified over-represented functions associated with oxidative stress, cell cycle, lipid metabolism, and immune responses consistent with the reported effects on redox status and histopathology at corresponding SDD drinking water concentrations. Collectively, these data are consistent with a mode of action involving oxidative stress and cytotoxicity as early key events. This suggests that the tumorigenic effects of chronic Cr(VI) oral exposure likely require chronic tissue damage and compensatory epithelial cell proliferation.


Asunto(s)
Cromo/toxicidad , Expresión Génica/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Contaminantes Químicos del Agua/toxicidad , Animales , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Agua Potable/química , Duodeno/efectos de los fármacos , Duodeno/metabolismo , Duodeno/patología , Femenino , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Yeyuno/efectos de los fármacos , Yeyuno/metabolismo , Yeyuno/patología , Ratones , Ratones Endogámicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Tiempo
20.
Regul Toxicol Pharmacol ; 64(1): 68-76, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22705708

RESUMEN

In vitro studies on hexavalent chromium [Cr(VI)] indicate that reduced forms of this metal can interact with DNA and cause mutations. Recently, Cr(VI) was shown to induce intestinal tumors in mice; however, Cr(VI) elicited redox changes, cytotoxicity and hyperplasia - suggesting involvement of tissue injury rather than direct mutagenesis. Moreover, toxicogenomic analyses indicated limited evidence for DNA damage responses. Herein, we extend these toxicogenomic analyses by comparing the gene expression patterns elicited by Cr(VI) with those of four mutagenic and four nonmutagenic carcinogens. To date, toxicogenomic profiles for mutagenic and nonmutagenic duodenal carcinogens do not exist, thus duodenal gene changes in mice were compared to those elicited by hepatocarcinogens. Specifically, duodenal gene changes in mice following exposure to Cr(VI) in drinking water were compared to hepatic gene changes previously identified as potentially discriminating mutagenic and nonmutagenic hepatocarcinogens. Using multivariate statistical analyses (including logistic regression classification), the Cr(VI) gene responses clustered apart from mutagenic carcinogens and closely with nonmutagenic carcinogens. These findings are consistent with other intestinal data supporting a nonmutagenic mode of action (MOA). These findings may be useful as part of a full weight of evidence MOA evaluation for Cr(VI)-induced intestinal carcinogenesis. Limitations to this analysis will also be discussed.


Asunto(s)
Carcinógenos Ambientales/toxicidad , Cromo/toxicidad , Mutágenos/toxicidad , Toxicogenética/métodos , Animales , Carcinógenos Ambientales/clasificación , Cromatos/clasificación , Cromatos/toxicidad , Cromo/clasificación , Simulación por Computador , Daño del ADN , Duodeno/efectos de los fármacos , Duodeno/metabolismo , Duodeno/patología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Estudio de Asociación del Genoma Completo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Modelos Logísticos , Ratones , Ratones Endogámicos , Pruebas de Mutagenicidad , Mutágenos/clasificación , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis de Componente Principal , Medición de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA