Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Environ Int ; 190: 108838, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38963985

RESUMEN

Known as "forever chemicals", per- and polyfluoroalkyl substances (PFAS) are synthetic compounds used in consumer goods but pose significant public health concerns, including disruption of the thyroid system. As thyroid hormones (THs) are required for normal brain development, PFAS may also be developmental neurotoxicants. However, this is not well understood. Here we examine the endocrine and neurodevelopmental consequences of perfluorohexane sulfonate (PFHxS) exposure in pregnant, lactating, and developing rats, and compare its effects to an anti-thyroid pharmaceutical (propylthiouracil, PTU) that induces thyroid-mediated developmental neurotoxicity. We show that PFHxS dramatically reduces maternal serum thyroxine (T4), nearly equivalently to PTU (-55 and -51%, respectively). However, only PTU increases thyroid stimulating hormone. The lactational transfer of PFHxS is significant and reduces pup serum T4 across the postnatal period. Surprisingly, brain THs are only minimally decreased by PFHxS, whereas PTU drastically diminishes them. Evaluation of brain TH action by phenotyping, RNA-Sequencing, and quantification of radial glia cell morphology supports that PTU interrupts TH signaling while PFHxS has limited to no effect. These data show that PFHxS induces abnormal serum TH profiles; however, there were no indications of hypothyroidism in the postnatal brain. We suggest the stark differences between the neurodevelopmental effects of PFHxS and a typical antithyroid agent may be due to its interaction with TH distributing proteins like transthyretin.

2.
Toxicol Sci ; 183(1): 195-213, 2021 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-34460931

RESUMEN

Many xenobiotics are identified as potential thyroid disruptors due to their action to reduce circulating levels of thyroid hormone, most notably thyroxine (T4). Developmental neurotoxicity is a primary concern for thyroid disrupting chemicals yet correlating the impact of chemically induced changes in serum T4 to perturbed brain development remains elusive. A number of thyroid-specific neurodevelopmental assays have been proposed, based largely on the model thyroid hormone synthesis inhibitor propylthiouracil (PTU). This study examined whether thyroid disrupting chemicals acting distinct from synthesis inhibition would result in the same alterations in brain as expected with PTU. The perfluoroalkyl substance perfluorohexane sulfonate (50 mg/kg/day) and the antimicrobial Triclosan (300 mg/kg/day) were administered to pregnant rats from gestational day 6 to postnatal day (PN) 21, and a number of PTU-defined assays for neurotoxicity evaluated. Both chemicals reduced serum T4 but did not increase thyroid stimulating hormone. Both chemicals increased expression of hepatic metabolism genes, while thyroid hormone-responsive genes in the liver, thyroid gland, and brain were largely unchanged. Brain tissue T4 was reduced in newborns, but despite persistent T4 reductions in serum, had recovered in the PN6 pup brain. Neither treatment resulted in a low dose PTU-like phenotype in either brain morphology or neurobehavior, raising questions for the interpretation of serum biomarkers in regulatory toxicology. They further suggest that reliance on serum hormones as prescriptive of specific neurodevelopmental outcomes may be too simplistic and to understand thyroid-mediated neurotoxicity we must expand our thinking beyond that which follows thyroid hormone synthesis inhibition.


Asunto(s)
Fluorocarburos , Triclosán , Animales , Femenino , Fluorocarburos/toxicidad , Embarazo , Propiltiouracilo/toxicidad , Ratas , Glándula Tiroides , Tiroxina , Triclosán/toxicidad
3.
Toxicol Pathol ; 36(4): 592-607, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18467677

RESUMEN

Health concerns have been raised because perfluorooctanoic acid (PFOA) is commonly found in the environment and can be detected in humans. In rodents, PFOA is a carcinogen and a developmental toxicant. PFOA is a peroxisome proliferator-activated receptor alpha (PPARalpha) activator; however, PFOA is capable of inducing heptomegaly in the PPARalpha-null mouse. To study the mechanism associated with PFOA toxicity, wild-type and PPARalpha-null mice were orally dosed for 7 days with PFOA (1 or 3 mg/kg) or the PPARalpha agonist Wy14,643 (50 mg/kg). Gene expression was evaluated using commercial microarrays. In wild-type mice, PFOA and Wy14,643 induced changes consistent with activation of PPARalpha. PFOA-treated wild-type mice deviated from Wy14,643-exposed mice with respect to genes involved in xenobiotic metabolism. In PFOA-treated null mice, changes were observed in transcripts related to fatty acid metabolism, inflammation, xenobiotic metabolism, and cell cycle regulation. Hence, a component of the PFOA response was found to be independent of PPARalpha. Although the signaling pathways responsible for these effects are not readily apparent, overlapping gene regulation by additional PPAR isoforms could account for changes related to fatty acid metabolism and inflammation, whereas regulation of xenobiotic metabolizing genes is suggestive of constitutive androstane receptor activation.


Asunto(s)
Caprilatos/toxicidad , Contaminantes Ambientales/toxicidad , Fluorocarburos/toxicidad , Perfilación de la Expresión Génica , Expresión Génica/efectos de los fármacos , Hígado/efectos de los fármacos , PPAR alfa/metabolismo , Animales , Caprilatos/farmacocinética , Contaminantes Ambientales/farmacocinética , Fluorocarburos/farmacocinética , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , PPAR alfa/genética , Pirimidinas/toxicidad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
Toxicol Sci ; 166(1): 163-179, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30085217

RESUMEN

Adverse neurodevelopmental consequences remain a primary concern when evaluating the effects of thyroid hormone (TH) disrupting chemicals. Though the developing brain is a known target of TH insufficiency, the relationship between THs in the serum and the central nervous system is not well characterized. To address this issue, dose response experiments were performed in pregnant rats using the goitrogen propylthiouracil (PTU) (dose range 0.1-10 ppm). THs were quantified in the serum and brain of offspring at gestational day 20 (GD20) and postnatal day 14 (PN14), two developmental stages included in OECD and EPA regulatory guideline/guidance studies. From the dose response data, the quantitative relationships between THs in the serum and brain were determined. Next, targeted gene expression analyses were performed in the fetal and neonatal cortex to test the hypothesis that TH action in the developing brain is linked to changes in TH concentrations within the tissue. Results show a significant reduction of T4/T3 in the serum and brain of the GD20 fetus in response to low doses of PTU; interestingly, very few genes were significantly different at any dose tested. In the PN14 pup significant reductions of T4/T3 in the serum and brain were also detected; however, twelve transcriptional targets were identified in the neonatal cortex that correlated well with reduced brain THs. These results show that serum T4 is a good predictor of brain THs, and offer several target genes that could serve as pragmatic readouts of T4/T3 dysfunction within the PN14 cortex.


Asunto(s)
Corteza Cerebral/metabolismo , Hipotiroidismo Congénito/metabolismo , Feto/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Hormonas Tiroideas/metabolismo , Animales , Animales Recién Nacidos , Antitiroideos/administración & dosificación , Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Hipotiroidismo Congénito/sangre , Hipotiroidismo Congénito/genética , Relación Dosis-Respuesta a Droga , Femenino , Feto/embriología , Expresión Génica/efectos de los fármacos , Exposición Materna/efectos adversos , Embarazo , Efectos Tardíos de la Exposición Prenatal/sangre , Efectos Tardíos de la Exposición Prenatal/genética , Propiltiouracilo/administración & dosificación , Ratas , Ratas Long-Evans , Hormonas Tiroideas/sangre
5.
Toxicol Sci ; 97(2): 595-613, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17383973

RESUMEN

Toxicogenomic analysis of five environmental chemicals was performed to investigate the ability of genomics to predict toxicity, categorize chemicals, and elucidate mechanisms of toxicity. Three triazole antifungals (myclobutanil, propiconazole, and triadimefon) and two perfluorinated chemicals [perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS)] were administered daily via oral gavage for one, three, or five consecutive days to male Sprague-Dawley rats at single doses of 300, 300, 175, 20, or 10 mg/kg/day, respectively. Clinical chemistry, hematology, and histopathology were measured at all time points. Gene expression profiling of livers from three rats per treatment group at all time points was performed on the CodeLink Uniset Rat I Expression array. Data were analyzed in the context of a large reference toxicogenomic database containing gene expression profiles for over 630 chemicals. Genomic signatures predicting hepatomegaly and hepatic injury preceded those results for all five chemicals, and further analysis segregated chemicals into two distinct classes. The triazoles caused similar gene expression changes as other azole antifungals, particularly the induction of pregnane X receptor (PXR)-regulated xenobiotic metabolism and oxidative stress genes. In contrast, PFOA and PFOS exhibited peroxisome proliferator-activated receptor alpha agonist-like effects on genes associated with fatty acid homeostasis. PFOA and PFOS also resulted in downregulation of cholesterol biosynthesis genes, matching an in vivo decrease in serum cholesterol, and perturbation of thyroid hormone metabolism genes matched by serum thyroid hormone depletion in vivo. The concordance of in vivo observations and gene expression findings demonstrated the ability of genomics to accurately categorize chemicals, identify toxic mechanisms of action, and predict subsequent pathological responses.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Fluorocarburos/toxicidad , Fungicidas Industriales/toxicidad , Hígado/metabolismo , Triazoles/toxicidad , Animales , Biomarcadores , Fluorocarburos/farmacocinética , Hormonas/sangre , Hígado/patología , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Tamaño de los Órganos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Testosterona/sangre , Hormonas Tiroideas/sangre , Triazoles/farmacocinética , Aumento de Peso/efectos de los fármacos
6.
Toxicology ; 239(1-2): 15-33, 2007 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-17681415

RESUMEN

Perfluorooctanoic acid (PFOA) is a stable perfluoroalkyl acid used to synthesize fluoropolymers during the manufacture of a wide variety of products. Concerns have been raised over the potential health effects of PFOA because it is persistent in the environment and can be detected in blood and other tissues of many animal species, including humans. PFOA has also been shown to induce growth deficits and mortality in murine neonates. To better understand the mechanism of PFOA induced developmental toxicity, lung and liver gene expression profiling was conducted in PFOA-exposed full-term mouse fetuses. Thirty timed-pregnant CD-1 mice were orally dosed from gestation days 1-17 with either 0, 1, 3, 5, or 10mg/(kgday) PFOA in water. At term, fetal lung and liver were collected, total RNA prepared, and samples pooled from three fetuses per litter. Five biological replicates consisting of individual litter samples were then evaluated for each treatment group using Affymetrix mouse 430_2 microarrays. The expression of genes related to fatty acid catabolism was altered in both the fetal liver and lung. In the fetal liver, the effects of PFOA were robust and also included genes associated with lipid transport, ketogenesis, glucose metabolism, lipoprotein metabolism, cholesterol biosynthesis, steroid metabolism, bile acid biosynthesis, phospholipid metabolism, retinol metabolism, proteosome activation, and inflammation. These changes are consistent with transactivation of PPARalpha, although, with regard to bile acid biosynthesis and glucose metabolism, non-PPARalpha related effects were suggested as well. Additional studies will be needed to more thoroughly address the role of PPARalpha, and other nuclear receptors, in PFOA mediated developmental toxicity.


Asunto(s)
Caprilatos/toxicidad , Contaminantes Ambientales/toxicidad , Desarrollo Fetal/efectos de los fármacos , Fluorocarburos/toxicidad , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hígado/efectos de los fármacos , Pulmón/efectos de los fármacos , Administración Oral , Animales , Relación Dosis-Respuesta a Droga , Ácidos Grasos/metabolismo , Femenino , Desarrollo Fetal/fisiología , Hígado/embriología , Hígado/metabolismo , Pulmón/embriología , Pulmón/metabolismo , Exposición Materna , Ratones , Ratones Endogámicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Embarazo , ARN Mensajero/metabolismo
7.
Toxicology ; 378: 37-52, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28049043

RESUMEN

Persistent presence of perfluoroalkyl acids (PFAAs) in the environment is due to their extensive use in industrial and consumer products, and their slow decay. Biochemical tests in rodent demonstrated that these chemicals are potent modifiers of lipid metabolism and cause hepatocellular steatosis. However, the molecular mechanism of PFAAs interference with lipid metabolism remains to be elucidated. Currently, two major hypotheses are that PFAAs interfere with mitochondrial beta-oxidation of fatty acids and/or they affect the transcriptional activity of peroxisome proliferator-activated receptor α (PPARα) in liver. To determine the ability of structurally-diverse PFAAs to cause steatosis, as well as to understand the underlying molecular mechanisms, wild-type (WT) and PPARα-null mice were treated with perfluorooctanoic acid (PFOA), perfluorononanoic acid (PFNA), or perfluorohexane sulfonate (PFHxS), by oral gavage for 7days, and their effects were compared to that of PPARα agonist WY-14643 (WY), which does not cause steatosis. Increases in liver weight and cell size, and decreases in DNA content per mg of liver, were observed for all compounds in WT mice, and were also seen in PPARα-null mice for PFOA, PFNA, and PFHxS, but not for WY. In Oil Red O stained sections, WT liver showed increased lipid accumulation in all treatment groups, whereas in PPARα-null livers, accumulation was observed after PFNA and PFHxS treatment, adding to the burden of steatosis observed in control (untreated) PPARα-null mice. Liver triglyceride (TG) levels were elevated in WT mice by all PFAAs and in PPARα-null mice only by PFNA. In vitro ß-oxidation of palmitoyl carnitine by isolated rat liver mitochondria was not inhibited by any of the 7 PFAAs tested. Likewise, neither PFOA nor PFOS inhibited palmitate oxidation by HepG2/C3A human liver cell cultures. Microarray analysis of livers from PFAAs-treated mice indicated that the PFAAs induce the expression of the lipid catabolism genes, as well as those involved in fatty acid and triglyceride synthesis, in WT mice and, to a lesser extent, in PPARα-null mice. These results indicate that most of the PFAAs increase liver TG load and promote steatosis in mice We hypothesize that PFAAs increase steatosis because the balance of fatty acid accumulation/synthesis and oxidation is disrupted to favor accumulation.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Contaminantes Ambientales/toxicidad , Hígado Graso/inducido químicamente , Hígado Graso/genética , Fluorocarburos/toxicidad , Metabolismo de los Lípidos/genética , Animales , Línea Celular Tumoral , ADN/metabolismo , Ácidos Grasos/metabolismo , Hígado Graso/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Homeostasis/efectos de los fármacos , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Mitocondrias Hepáticas/fisiología , PPAR alfa/genética , Palmitatos/metabolismo , Ratas Sprague-Dawley , Triglicéridos/metabolismo
9.
Reprod Toxicol ; 19(3): 353-66, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15686870

RESUMEN

A byproduct of drinking water disinfection, bromochloroacetic acid (BCA), acts as a reproductive toxicant in rats. To determine if BCA produces similar reproductive toxicity in mice, juvenile and adult C57BL/6 males were exposed to 0, 8, 24, 72 or 216 mg/kg of BCA once daily for 14 days. Five of 12 animals from each dose-group were sacrificed at the end of dosing, and testes, epididymes, and seminal vesicles harvested and weighed. Seven mice from each dose-group (including juvenile-exposed mice, following a 14-week maturation period) were used in a 40-day sequential breeding assay to determine if BCA targets a particular phase of spermatogenesis. No significant effects were observed in mice exposed to BCA as juveniles, and there were no effects on fertility by 14 weeks after dosing. However, effects were observed in adult-exposed mice over the first 10 days after BCA exposure: mean number of litters/male, percentage of litters/female bred, and total number of fetuses/male were all reduced by 72 and 216 mg/kg BCA. These results in adult mice indicate BCA disrupted differentiation of spermatids during dosing and the first 10 days of mating, and are consistent with the spermatid retention and atypical residual bodies observed in animals exposed to 72 and 216 mg/kg BCA. To investigate mechanisms involved, we utilized cDNA microarrays containing 950 testis-expressed genes to profile gene expression from Control and BCA-treated mice. Statistical analyses of microarray results identified 40 well-characterized genes differentially expressed in a dose responsive manner as a result of BCA exposure. Microarray results were supplemented with quantitative real-time PCR and Westerns for several genes and proteins. The 40 genes whose expression was altered by BCA are involved in numerous biological processes including: cell communication and adhesion, cell cycle and cell proliferation, metabolism, signal transduction, stress response, and spermatogenesis and male fertility. Modulated expression of these genes, particularly the 15 expressed in Sertoli cells and spermatids, offers new insights into potential mechanisms of BCA toxicity in the mouse testis.


Asunto(s)
Acetatos/toxicidad , Desinfectantes/toxicidad , Testículo/efectos de los fármacos , Animales , Western Blotting , Diferenciación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Células de Sertoli/efectos de los fármacos , Espermátides/efectos de los fármacos , Testículo/patología , Pruebas de Toxicidad Aguda
10.
Mol Cell Endocrinol ; 400: 90-101, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25448844

RESUMEN

The 3T3-L1 preadipocyte culture system has been used to examine numerous compounds that influence adipocyte differentiation or function. The perfluoroalkyl acids (PFAAs), used as surfactants in a variety of industrial applications, are of concern as environmental contaminants that are detected worldwide in human serum and animal tissues. This study was designed to evaluate the potential for PFAAs to affect adipocyte differentiation and lipid accumulation using mouse 3T3-L1 cells. Cells were treated with perfluorooctanoic acid (PFOA) (5-100 µM), perfluorononanoic acid (PFNA) (5-100 µM), perfluorooctane sulfonate (PFOS) (50-300 µM), perfluorohexane sulfonate (PFHxS) (40-250 µM), the peroxisome proliferator activated receptor (PPAR) PPARα agonist Wyeth-14,643 (WY-14,643), and the PPARγ agonist rosiglitazone. The PPARγ agonist was included as a positive control as this pathway is critical to adipocyte differentiation. The PPARα agonist was included as the PFAA compounds are known activators of this pathway. Cells were assessed morphometrically and biochemically for number, size, and lipid content. RNA was extracted for qPCR analysis of 13 genes selected for their importance in adipocyte differentiation and lipid metabolism. There was a significant concentration-related increase in cell number and decreased cell size after exposure to PFOA, PFHxS, PFOS, and PFNA. All four PFAA treatments produced a concentration-related decrease in the calculated average area occupied by lipid per cell. However, total triglyceride levels per well increased with a concentration-related trend for all compounds, likely due to the increased cell number. Expression of mRNA for the selected genes was affected by all exposures and the specific impacts depended on the particular compound and concentration. Acox1 and Gapdh were upregulated by all six compounds. The strongest overall effect was a nearly 10-fold induction of Scd1 by PFHxS. The sulfonated PFAAs produced numerous, strong changes in gene expression similar to the effects after treatment with the PPARγ agonist rosiglitazone. By comparison, the effects on gene expression were muted for the carboxylated PFAAs and for the PPARα agonist WY-14,643. In summary, all perfluorinated compounds increased cell number, decreased cell size, increased total triglyceride, and altered expression of genes associated with adipocyte differentiation and lipid metabolism.


Asunto(s)
Adipocitos/efectos de los fármacos , Contaminantes Ambientales/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Estearoil-CoA Desaturasa/metabolismo , Células 3T3-L1 , Adipocitos/citología , Adipocitos/metabolismo , Ácidos Alcanesulfónicos/farmacología , Animales , Caprilatos/farmacología , Diferenciación Celular/efectos de los fármacos , Tamaño de la Célula , Ácidos Grasos , Fluorocarburos/farmacología , Perfilación de la Expresión Génica , Ratones , PPAR alfa/agonistas , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR gamma/agonistas , PPAR gamma/genética , PPAR gamma/metabolismo , Pirimidinas/farmacología , Rosiglitazona , Transducción de Señal , Estearoil-CoA Desaturasa/genética , Tiazolidinedionas/farmacología
11.
Reprod Toxicol ; 51: 133-44, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25543169

RESUMEN

Perfluorononanoic acid (PFNA) is a ubiquitous and persistent environmental contaminant. Although its levels in the environment and in humans are lower than those of perfluorooctane sulfonate (PFOS) or perfluorooctanoic acid (PFOA), a steady trend of increases in the general population in recent years has drawn considerable interest and concern. Previous studies with PFOS and PFOA have indicated developmental toxicity in laboratory rodent models. The current study extends the evaluation of these adverse outcomes to PFNA in mice. PFNA was given to timed-pregnant CD-1 mice by oral gavage daily on gestational day 1-17 at 1, 3, 5 or 10mg/kg; controls received water vehicle. Dams given 10mg/kg PFNA could not carry their pregnancy successfully and effects of this dose group were not followed. Similar to PFOS and PFOA, PFNA at 5mg/kg or lower doses produced hepatomegaly in the pregnant dams, but did not affect the number of implantations, fetal viability, or fetal weight. Mouse pups were born alive and postnatal survival in the 1 and 3mg/kg PFNA groups was not different from that in controls. In contrast, although most of the pups were also born alive in the 5mg/kg PFNA group, 80% of these neonates died in the first 10 days of life. The pattern of PFNA-induced neonatal death differed somewhat from those elicited by PFOS or PFOA. A majority of the PFNA-exposed pups survived a few days longer after birth than those exposed to PFOS or PFOA, which typically died within the first 2 days of postnatal life. Surviving neonates exposed to PFNA exhibited dose-dependent delays in eye opening and onset of puberty. In addition, increased liver weight seen in PFNA-exposed offspring persisted into adulthood and was likely related to the persistence of the chemical in the tissue. Evaluation of gene expression in fetal and neonatal livers revealed robust activation of peroxisome proliferator-activated receptor-alpha (PPARα) target genes by PFNA that resembled the responses of PFOA. Our results indicate that developmental toxicity of PFNA in mice is comparable to that of PFOS and PFOA, and that these adverse effects are likely common to perfluoroalkyl acids that persist in the body.


Asunto(s)
Fluorocarburos/toxicidad , Hígado/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Ácidos Grasos , Femenino , Fluorocarburos/sangre , Fluorocarburos/farmacocinética , Hígado/metabolismo , Hígado/patología , Intercambio Materno-Fetal , Ratones , Tamaño de los Órganos/efectos de los fármacos , PPAR alfa/genética , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Transcriptoma
12.
Toxicol Sci ; 139(1): 21-34, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24496636

RESUMEN

More efficient models are needed to assess potential carcinogenicity hazard of environmental chemicals based on early events in tumorigenesis. Here, we investigated time course profiles for key events in an established cancer mode of action. Using a case study approach, we evaluated two reference phthalates, di(2-ethylhexyl) phthalate (DEHP) and its stereoisomer di-n-octyl phthalate (DNOP), across the span of a two-year carcinogenicity bioassay. Male B6C3F1 mice received diets with no phthalate added (control), DEHP at 0.12, 0.60, or 1.20%, or DNOP at 0.10, 0.50, or 1.00% (n = 80-83/group) for up to 104 weeks with six interim evaluations starting at week 4. Mean phthalate doses were 139, 845, and 3147 mg/kg/day for DEHP and 113, 755, and 1281 mg/kg/day for DNOP groups, respectively. Incidence and number of hepatocellular tumors (adenoma and/or carcinoma) were greater at ≥ 60 weeks for all DEHP groups with time and dose trends, whereas DNOP had no significant effects. Key events supported a peroxisome proliferator-activated receptor alpha (PPARα) mode of action for DEHP, with secondary cytotoxicity at the high dose, whereas DNOP induced modest increases in PPARα activity without proliferative or cytotoxic effects. Threshold estimates for later tumorigenic effects were identified at week 4 for relative liver weight (+24%) and PPARα activity (+79%) relative to the control group. Benchmark doses (BMDs) for these measures at week 4 clearly distinguished DEHP and DNOP and showed strong concordance with values at later time points and tumorigenic BMDs. Other target sites included testis and kidney, which showed degenerative changes at higher doses of DEHP but not DNOP. Our results highlight marked differences in the chronic toxicity profiles of structurally similar phthalates and demonstrate quantitative relationships between early bioindicators and later tumor outcomes.


Asunto(s)
Ácidos Ftálicos/toxicidad , Animales , Carcinogénesis , Relación Dosis-Respuesta a Droga , Hígado/efectos de los fármacos , Hígado/metabolismo , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/metabolismo , Masculino , Ratones , PPAR alfa/metabolismo , Ácidos Ftálicos/administración & dosificación , Ácidos Ftálicos/química , Estereoisomerismo
13.
Toxicology ; 308: 129-37, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23567314

RESUMEN

While perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS) have been studied at length, less is known about the biological activity of other perfluoroalkyl acids (PFAAs) detected in the environment. Using a transient transfection assay developed in COS-1 cells, our group has previously evaluated a variety of PFAAs for activity associated with activation of peroxisome proliferator-activated receptor alpha (PPARα). Here we use primary heptatocytes to further assess the biological activity of a similar group of PFAAs using custom designed Taqman Low Density Arrays. Primary mouse and human hepatoyctes were cultured for 48h in the presence of varying concentrations of 12 different PFAAs or Wy14,643, a known activator of PPARα. Total RNA was collected and the expression of 48 mouse or human genes evaluated. Gene selection was based on either in-house liver microarray data (mouse) or published data using primary hepatocytes (human). Gene expression in primary mouse hepatocytes was more restricted than expected. Genes typically regulated in whole tissue by PPARα agonists were not altered in mouse cells including Acox1, Me1, Acaa1a, Hmgcs1, and Slc27a1. Cyp2b10, a gene regulated by the constitutive androstane receptor and a transcript normally up-regulated by in vivo exposure to PFAAs, was also unchanged in cultured mouse hepatocytes. Cyp4a14, Ehhadh, Pdk4, Cpt1b, and Fabp1 were regulated as expected in mouse cells. A larger group of genes were differentially expressed in human primary hepatocytes, however, little consistency was observed across compounds with respect to which genes produced a significant dose response making the determination of relative biological activity difficult. This likely reflects weaker activation of PPARα in human versus rodent cells as well as variation among individual cell donors. Unlike mouse cells, CYP2B6 was up-regulated in human hepatocytes by a number of PFAAs as was PPARδ. Rankings were conducted on the limited dataset. In mouse hepatocytes, the pattern was similar to that previously observed in the COS-1 reporter cell assay. With the exception of PFHxA, longer chain PFAA carboxylates were the most active. The pattern was similar in human hepatocytes, although PFDA and PFOS showed higher activity than previously observed while PFOA showed somewhat less activity. These data reflect inherent challenges in using primary hepatocytes to predict toxicological response.


Asunto(s)
Ácidos Alcanesulfónicos/química , Ácidos Alcanesulfónicos/toxicidad , Fluorocarburos/química , Fluorocarburos/toxicidad , Hepatocitos/efectos de los fármacos , Animales , Evaluación Preclínica de Medicamentos/métodos , Humanos , Ratones , Cultivo Primario de Células
14.
Reprod Toxicol ; 33(4): 491-505, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22154759

RESUMEN

PPARs regulate metabolism and can be activated by environmental contaminants such as perfluorooctanoic acid (PFOA). PFOA induces neonatal mortality, developmental delay, and growth deficits in mice. Studies in genetically altered mice showed that PPARα is required for PFOA-induced developmental toxicity. In this study, pregnant CD-1 mice were dosed orally from GD1 to 17 with water or 5mg PFOA/kg to examine PPARα, PPARß, and PPARγ expression and profile the effects of PFOA on PPAR-regulated genes. Prenatal and postnatal liver, heart, adrenal, kidney, intestine, stomach, lung, spleen, and thymus were collected at various developmental ages. RNA and protein were examined using qPCR and Western blot analysis. PPAR expression varied with age in all tissues, and in liver PPARα and PPARγ expression correlated with nutritional changes as the pups matured. As early as GD14, PFOA affected expression of genes involved in lipid and glucose homeostatic control. The metabolic disruption produced by PFOA may contribute to poor postnatal survival and persistent weight deficits of CD-1 mouse neonates.


Asunto(s)
Caprilatos/toxicidad , Contaminantes Ambientales/toxicidad , Desarrollo Fetal , Fluorocarburos/toxicidad , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Receptores Activados del Proliferador del Peroxisoma/genética , Efectos Tardíos de la Exposición Prenatal , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Animales Recién Nacidos , Western Blotting , Caprilatos/sangre , Caprilatos/farmacocinética , Contaminantes Ambientales/sangre , Contaminantes Ambientales/farmacocinética , Femenino , Desarrollo Fetal/efectos de los fármacos , Desarrollo Fetal/genética , Fluorocarburos/sangre , Fluorocarburos/farmacocinética , Edad Gestacional , Masculino , Ratones , Ratones Endogámicos , Especificidad de Órganos , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
15.
PPAR Res ; 20102010.
Artículo en Inglés | MEDLINE | ID: mdl-20706641

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) regulate lipid and glucose homeostasis, are targets of pharmaceuticals, and are also activated by environmental contaminants. Almost nothing is known about expression of PPARs during human fetal development. This study examines expression of PPARalpha, beta, and gamma mRNA and protein in human fetal tissues. With increasing fetal age, mRNA expression of PPARalpha and beta increased in liver, but PPARbeta decreased in heart and intestine, and PPARgamma decreased in adrenal. Adult and fetal mean expression of PPARalpha, beta, and gamma mRNA did not differ in intestine, but expression was lower in fetal stomach and heart. PPARalpha and beta mRNA in kidney and spleen, and PPARgamma mRNA in lung and adrenal were lower in fetal versus adult. PPARgamma in liver and PPARbeta mRNA in thymus were higher in fetal versus adult. PPARalpha protein increased with fetal age in intestine and decreased in lung, kidney, and adrenal. PPARbeta protein in adrenal and PPARgamma in kidney decreased with fetal age. This study provides new information on expression of PPAR subtypes during human development and will be important in evaluating the potential for the developing human to respond to PPAR environmental or pharmaceutical agonists.

16.
Reprod Toxicol ; 27(3-4): 278-288, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19429403

RESUMEN

Perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) are environmental contaminants found in the tissues of humans and wildlife. They are activators of peroxisome proliferator-activated receptor-alpha (PPAR alpha) and exhibit hepatocarcinogenic potential in rats. PFOS and PFOA are also developmental toxicants in rodents and PFOS has been shown to induce pulmonary deficits in rat offspring. Pregnant CD-1 mice were dosed with 0, 5, or 10mg/kg PFOS from gestation days 1-17. Transcript profiling was conducted on the fetal liver and lung. Results were contrasted to data derived from a previous PFOA study. PFOS-dependent changes were primarily related to activation of PPAR alpha. No remarkable differences were found between PFOS and PFOA. Given that PPAR alpha signaling is required for neonatal mortality in PFOA-treated mice but not those exposed to PFOS, the neonatal mortality observed for PFOS may reflect functional deficits related to the physical properties of the chemical rather than to transcript alterations.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Caprilatos/toxicidad , Fluorocarburos/toxicidad , Perfilación de la Expresión Génica , Hígado/metabolismo , Pulmón/metabolismo , Ácidos Alcanesulfónicos/farmacología , Animales , Caprilatos/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Feto/metabolismo , Fluorocarburos/farmacología , Exposición Materna , Ratones , Ratones Endogámicos , Análisis por Micromatrices , Embarazo
17.
Brain Res Bull ; 76(5): 531-5, 2008 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-18534262

RESUMEN

Glucocorticoids are the consensus treatment to avoid respiratory distress in preterm infants but there is accumulating evidence that these agents evoke long-term neurobehavioral deficits. Earlier, we showed that the developing rat forebrain is far more sensitive to glucocorticoid-induced disruption in the fetus than in the neonate. Feedback regulation of glucocorticoid receptors (GRs) is an essential homeostatic mechanism and we therefore examined the development of GR downregulation in the perinatal period. Pregnant rats or newborn pups were given dexamethasone daily (gestational days 17-19, postnatal days 1-3, or postnatal days 7-9), ranging from doses below that recommended for use in preterm infants (0.05 mg/kg) to therapeutic doses (0.2 or 0.8 mg/kg). Twenty-four hours after the last injection, we determined forebrain GR protein by Western blotting. Although postnatal dexamethasone treatment downregulated GRs at all doses, the fetal forebrain failed to show any decrement and instead exhibited slight GR upregulation. In controls, forebrain GR levels also showed a large increment over the course from late gestation through the second postnatal week, despite the fact that circulating glucocorticoid levels increase substantially during this period. Our results suggest that GR homeostasis develops primarily postnatally and that fetal inability to downregulate GRs in the face of exogenous glucocorticoid administration plays a role in the vulnerability of key neural circuits to developmental disruption. Since this developmental phase in the rat corresponds to the critical period in which glucocorticoids are used in preterm infants, adverse effects on brain development may be inescapable.


Asunto(s)
Dexametasona , Recien Nacido Prematuro , Prosencéfalo , Receptores de Glucocorticoides/fisiología , Animales , Dexametasona/metabolismo , Dexametasona/farmacología , Femenino , Homeostasis , Humanos , Recién Nacido , Masculino , Embarazo , Prosencéfalo/efectos de los fármacos , Prosencéfalo/crecimiento & desarrollo , Prosencéfalo/metabolismo , Ratas , Ratas Sprague-Dawley
18.
Toxicol Sci ; 105(1): 173-81, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18511431

RESUMEN

Perfluorobutyrate (PFBA) is a perfluoroalkyl acid (PFAA) found in the environment. Previous studies have indicated developmental toxicity of PFAAs (perfluorooctane sulfonate [PFOS] and perfluorooctanoate [PFOA]); the current study examines that of PFBA. PFBA/NH4(+) was given to timed-pregnant CD-1 mice by oral gavage daily from gestational day (GD) 1 to 17 at 35, 175, or 350 mg/kg (chosen to approximate the developmentally toxic doses of PFOA); controls received water. At GD 18, serum levels of PFBA were 3.8, 4.4, and 2.5 microg/ml, respectively, in the three treated groups. PFBA did not significantly affect maternal weight gain, number of implantations, fetal viability, fetus weight, or incidence of fetal malformations. Incidence of full-litter loss was significantly greater in the 350 mg/kg group, and maternal liver weights were significantly increased in the 175 and 350 mg/kg groups. In contrast to PFOA and PFOS, PFBA exposure during pregnancy did not adversely affect neonatal survival or postnatal growth. Liver enlargement was detected in the PFBA-exposed pups on postnatal day (PD) 1, but not by PD 10. Expression of selected hepatic genes in PFBA-exposed pups at PD 1 did not reveal any significant changes from controls. A significant delay in eye-opening in offspring was detected in all three PFBA groups, and slight delays in the onset of puberty were noted in the 175 and 350 mg/kg groups. These data suggest that exposure to PFBA during pregnancy in the mouse did not produce developmental toxicity comparable to that observed with PFOA, in part, due to rapid elimination of the chemical.


Asunto(s)
Feto/efectos de los fármacos , Fluorocarburos/toxicidad , Anomalías Inducidas por Medicamentos , Animales , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Fluorocarburos/sangre , Expresión Génica/efectos de los fármacos , Crecimiento/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Tamaño de los Órganos/efectos de los fármacos , Embarazo
19.
Toxicol Appl Pharmacol ; 215(3): 260-73, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16643972

RESUMEN

Four triazole fungicides were studied using toxicogenomic techniques to identify potential mechanisms of action. Adult male Sprague-Dawley rats were dosed for 14 days by gavage with fluconazole, myclobutanil, propiconazole, or triadimefon. Following exposure, serum was collected for hormone measurements, and liver and testes were collected for histology, enzyme biochemistry, or gene expression profiling. Body and testis weights were unaffected, but liver weights were significantly increased by all four triazoles, and hepatocytes exhibited centrilobular hypertrophy. Myclobutanil exposure increased serum testosterone and decreased sperm motility, but no treatment-related testis histopathology was observed. We hypothesized that gene expression profiles would identify potential mechanisms of toxicity and used DNA microarrays and quantitative real-time PCR (qPCR) to generate profiles. Triazole fungicides are designed to inhibit fungal cytochrome P450 (CYP) 51 enzyme but can also modulate the expression and function of mammalian CYP genes and enzymes. Triazoles affected the expression of numerous CYP genes in rat liver and testis, including multiple Cyp2c and Cyp3a isoforms as well as other xenobiotic metabolizing enzyme (XME) and transporter genes. For some genes, such as Ces2 and Udpgtr2, all four triazoles had similar effects on expression, suggesting possible common mechanisms of action. Many of these CYP, XME and transporter genes are regulated by xeno-sensing nuclear receptors, and hierarchical clustering of CAR/PXR-regulated genes demonstrated the similarities of toxicogenomic responses in liver between all four triazoles and in testis between myclobutanil and triadimefon. Triazoles also affected expression of multiple genes involved in steroid hormone metabolism in the two tissues. Thus, gene expression profiles helped identify possible toxicological mechanisms of the triazole fungicides.


Asunto(s)
Antifúngicos/toxicidad , Fungicidas Industriales/toxicidad , Hígado/efectos de los fármacos , Testículo/efectos de los fármacos , Triazoles/toxicidad , Animales , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Ratas Sprague-Dawley , Motilidad Espermática/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Espermatozoides/fisiología , Testículo/metabolismo , Testosterona/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA