Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Toxicology ; 441: 152522, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32534104

RESUMO

Per- and polyfluoroalkyl substances (PFAS) are organic chemicals with wide industrial and consumer uses. They are found ubiquitously at low levels in the environment and are detectable in humans and wildlife. Perfluorobutane Sulfonate (PFBS) is a short-chained PFAS used to replace perfluorooctane sulfonate in commerce. In general, the rate of clearance for the short-chained PFAS is faster than that for the long-chained congeners. This study evaluated the pharmacokinetic properties of PFBS and its hepatic transcriptional responses in CD-1 mice. Males and females were given PFBS by oral gavage at 30 or 300 mg/kg; controls received 0.5 % Tween-20 vehicle. Trunk blood was collected at 0.5, 1, 2, 4, 8, 16 and 24 h thereafter; liver and kidney were also harvested. Serum and tissue concentrations of PFBS were determined by HPLC-MS-MS. Expression of several hepatic nuclear receptor target genes was determined by qPCR. The half-life of PFBS was estimated as 5.8 h in the males and 4.5 h in the females. Tmax was reached within 1-2 h. Volume of distribution was similar between the two sexes (0.32-0.40 L/kg). The rate of PFBS clearance was linear with exposure doses. Within 24 h, serum PFBS declined to less than 5 % of Cmax. PFBS was detected in liver or kidney, although tissue levels of the chemical were only a fraction of those in serum. At 24 h after administration of 300 mg/kg PFBS, elevated expression of several hepatic genes targeted for PPARα, PPARy, and PXR but not by AhR, LXR or CAR was observed, with responses indistinguishable between males and females. Little to no transcriptional response was seen with the 30 mg/kg dose. The short serum half-lives of PFBS (4-5 h) in mice were comparable to those reported in rats. Although detection of PFBS in liver was low compared to that in serum even at the 300 mg/kg dose, the tissue level was sufficient to activate several hepatic nuclear receptors, which may represent an acute response to the chemical at a high dose.


Assuntos
Fluorocarbonos/farmacocinética , Fígado/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Ácidos Sulfônicos/farmacocinética , Animais , Relação Dose-Resposta a Droga , Feminino , Meia-Vida , Rim/efeitos dos fármacos , Rim/metabolismo , Fígado/metabolismo , Masculino , Taxa de Depuração Metabólica , Camundongos , Reação em Cadeia da Polimerase , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores Sexuais , Transcriptoma/efeitos dos fármacos
2.
Birth Defects Res ; 110(17): 1322-1334, 2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30347137

RESUMO

BACKGROUND: Cleft palate (CP) is a common birth defect, occurring in an estimated 1 in 1,000 births worldwide. The secondary palate is formed by paired palatal shelves, consisting of a mesenchymal core with an outer layer of epithelial cells that grow toward each other, attach, and fuse. One of the mechanisms that can cause CP is failure of fusion, that is, failure to remove the epithelial seam between the palatal shelves to allow the mesenchyme confluence. Epidermal growth factor (EGF) plays an important role in palate growth and differentiation, while it may impede fusion. METHODS: We developed a 3D organotypic model using human mesenchymal and epithelial stem cells to mimic human embryonic palatal shelves, and tested the effects of human EGF (hEGF) on proliferation and fusion. Spheroids were generated from human umbilical-derived mesenchymal stem cells (hMSCs) directed down an osteogenic lineage. Heterotypic spheroids, or organoids, were constructed by coating hMSC spheroids with extracellular matrix solution followed by a layer of human progenitor epithelial keratinocytes (hPEKs). Organoids were incubated in co-culture medium with or without hEGF and assessed for cell proliferation and time to fusion. RESULTS: Osteogenic differentiation in hMSC spheroids was highest by Day 13. hEGF delayed fusion of organoids after 12 and 18 hr of contact. hEGF increased proliferation in organoids at 4 ng/ml, and proliferation was detected in hPEKs alone. CONCLUSION: Our results show that this model of human palatal fusion appropriately mimics the morphology of the developing human palate and responds to hEGF as expected.


Assuntos
Desenvolvimento Ósseo/fisiologia , Fissura Palatina/embriologia , Fator de Crescimento Epidérmico/metabolismo , Células Epiteliais/citologia , Células-Tronco Mesenquimais/citologia , Palato/embriologia , Proliferação de Células/fisiologia , Células Cultivadas , Humanos , Osteogênese/fisiologia , Esferoides Celulares/citologia , Veias Umbilicais/citologia
3.
Toxicology ; 387: 95-107, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28558994

RESUMO

Perfluoroalkyl acids (PFAAs) are ubiquitous and persistent environmental contaminants. Compounds such as perfluoroocanoic acid (PFOA), perfluorooctane sulfonate (PFOS), perfluorononanoic acid (PFNA), and perfluorohexane sulfonate (PFHxS) are readily found in the tissues of humans and wildlife. While PFOA and PFOS have been the subject of numerous studies since they were first described over a decade ago, less is known about the biological activity of PFHxS and PFNA. Most PFAAs are activators of peroxisome proliferator-activated receptor α (PPARα), although the biological effects of these compounds are likely mediated by other factors in addition to PPARα. To evaluate the effects of PFHxS and PFNA, male wild-type and Pparα-null mice were dosed by oral gavage with PFHxS (3 or 10mg/kg/day), PFNA (1 or 3mg/kg/day), or vehicle for 7days, and liver gene expression was evaluated by full-genome microarrays. Gene expression patterns were then compared to historical in-house data for PFOA and PFOS in addition to the experimental hypolipidemic agent, WY-14,643. While WY-14,643 altered most genes in a PPARα-dependent manner, approximately 11-24% of regulated genes in PFAA-treated mice were independent of PPARα. The possibility that PFAAs regulate gene expression through other molecular pathways was evaluated. Using data available through a microarray database, PFAA gene expression profiles were found to exhibit significant similarity to profiles from mouse tissues exposed to agonists of the constitutive activated receptor (CAR), estrogen receptor α (ERα), and PPARγ. Human PPARγ and ERα were activated by all four PFAAs in trans-activation assays from the ToxCast screening program. Predictive gene expression biomarkers showed that PFAAs activate CAR in both genotypes and cause feminization of the liver transcriptome through suppression of signal transducer and activator of transcription 5B (STAT5B). These results indicate that, in addition to activating PPARα as a primary target, PFAAs also have the potential to activate CAR, PPARγ, and ERα as well as suppress STAT5B.


Assuntos
Fluorocarbonos/toxicidade , Perfilação da Expressão Gênica/métodos , Fígado/efeitos dos fármacos , Análise de Sequência com Séries de Oligonucleotídeos , PPAR alfa/agonistas , Ácidos Sulfônicos/toxicidade , Transcrição Gênica/efeitos dos fármacos , Animais , Anticolesterolemiantes/farmacologia , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Biologia Computacional , Receptor Constitutivo de Androstano , Bases de Dados Genéticas , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Estrogênios/farmacologia , Ácidos Graxos , Regulação da Expressão Gênica , Hepatomegalia/induzido quimicamente , Hepatomegalia/genética , Hepatomegalia/metabolismo , Hepatomegalia/patologia , Humanos , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos da Linhagem 129 , Camundongos Knockout , PPAR alfa/deficiência , PPAR alfa/genética , PPAR gama/agonistas , PPAR gama/genética , PPAR gama/metabolismo , Pirimidinas/farmacologia , Receptores Citoplasmáticos e Nucleares/agonistas , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/efeitos dos fármacos
4.
Toxicology ; 378: 37-52, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-28049043

RESUMO

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.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Poluentes Ambientais/toxicidade , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/genética , Fluorocarbonos/toxicidade , Metabolismo dos Lipídeos/genética , Animais , Linhagem Celular Tumoral , DNA/metabolismo , Ácidos Graxos/metabolismo , Fígado Gorduroso/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias Hepáticas/efeitos dos fármacos , Mitocôndrias Hepáticas/metabolismo , Mitocôndrias Hepáticas/fisiologia , PPAR alfa/genética , Palmitatos/metabolismo , Ratos Sprague-Dawley , Triglicerídeos/metabolismo
5.
Reprod Toxicol ; 51: 133-44, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25543169

RESUMO

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.


Assuntos
Fluorocarbonos/toxicidade , Fígado/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Ácidos Graxos , Feminino , Fluorocarbonos/sangue , Fluorocarbonos/farmacocinética , Fígado/metabolismo , Fígado/patologia , Troca Materno-Fetal , Camundongos , Tamanho do Órgão/efeitos dos fármacos , PPAR alfa/genética , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Transcriptoma
6.
Toxicology ; 308: 129-37, 2013 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-23567314

RESUMO

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.


Assuntos
Ácidos Alcanossulfônicos/química , Ácidos Alcanossulfônicos/toxicidade , Fluorocarbonos/química , Fluorocarbonos/toxicidade , Hepatócitos/efeitos dos fármacos , Animais , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Camundongos , Cultura Primária de Células
7.
Reprod Toxicol ; 33(4): 491-505, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22154759

RESUMO

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.


Assuntos
Caprilatos/toxicidade , Poluentes Ambientais/toxicidade , Desenvolvimento Fetal , Fluorocarbonos/toxicidade , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Receptores Ativados por Proliferador de Peroxissomo/genética , Efeitos Tardios da Exposição Pré-Natal , Envelhecimento/genética , Envelhecimento/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Caprilatos/sangue , Caprilatos/farmacocinética , Poluentes Ambientais/sangue , Poluentes Ambientais/farmacocinética , Feminino , Desenvolvimento Fetal/efeitos dos fármacos , Desenvolvimento Fetal/genética , Fluorocarbonos/sangue , Fluorocarbonos/farmacocinética , Idade Gestacional , Masculino , Camundongos , Camundongos Endogâmicos , Especificidade de Órgãos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
8.
Toxicology ; 281(1-3): 48-55, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21237237

RESUMO

Perfluorononanoic acid (PFNA) is a fluorinated organic chemical found at low levels in the environment, but is detectable in humans and wildlife. The present study compared the pharmacokinetic properties of PFNA in two laboratory rodent species. Male and female Sprague-Dawley rats were given a single dose of PFNA by oral gavage at 1, 3, or 10mg/kg, and blood was collected from the tail vein at 1, 2, 3, 4, 7, 16, 21, 28, 35, 42 and 50 days after treatment. In addition, livers and kidneys were collected for PFNA analysis at the terminal time point. CD-1 mice were given a single oral dose of PFNA of 1 or 10mg/kg, and 4 males and 4 females were killed at similar time intervals; trunk blood, liver and kidney were collected. Serum and tissue concentrations of PFNA were determined by LC-MS/MS. Serum elimination of PFNA is by and large linear with exposure doses in the rat; however, like PFOA, a major sex difference in the rate of elimination is observed, with an estimated half-life of 30.6 days for males and 1.4 days for females. PFNA is stored preferentially in the liver but not in the kidneys. In the mouse, the rates of PFNA serum elimination are non-linear with exposure dose and are slightly faster in females than males, with terminal estimated serum half-life of 25.8-68.4 days and 34.3-68.9 days, respectively. PFNA is also stored preferentially in the mouse liver but not in the kidneys. Hepatic uptake appears to be more efficient and storage capacity greater in male mice than in females. These data suggest that (1) PFNA is more persistent in the mouse than in the rat; (2) there is a major sex difference in the serum elimination of PFNA in the rat, but much less so in the mouse; and (3) there is a significantly higher hepatic accumulation of PFNA in male mice than in females.


Assuntos
Ácidos Graxos/farmacocinética , Hidrocarbonetos Fluorados/farmacocinética , Administração Oral , Animais , Ácidos Graxos/análise , Ácidos Graxos/sangue , Feminino , Fluorocarbonos , Cromatografia Gasosa-Espectrometria de Massas , Meia-Vida , Hidrocarbonetos Fluorados/análise , Hidrocarbonetos Fluorados/sangue , Rim/química , Fígado/química , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley , Fatores Sexuais
9.
PPAR Res ; 20102010.
Artigo em Inglês | MEDLINE | ID: mdl-20936131

RESUMO

Perfluorooctane sulfonate (PFOS) is a perfluoroalkyl acid (PFAA) and a persistent environmental contaminant found in the tissues of humans and wildlife. Although blood levels of PFOS have begun to decline, health concerns remain because of the long half-life of PFOS in humans. Like other PFAAs, such as, perfluorooctanoic acid (PFOA), PFOS is an activator of peroxisome proliferator-activated receptor-alpha (PPARα) and exhibits hepatocarcinogenic potential in rodents. PFOS is also a developmental toxicant in rodents where, unlike PFOA, its mode of action is independent of PPARα. Wild-type (WT) and PPARα-null (Null) mice were dosed with 0, 3, or 10 mg/kg/day PFOS for 7 days. Animals were euthanized, livers weighed, and liver samples collected for histology and preparation of total RNA. Gene profiling was conducted using Affymetrix 430_2 microarrays. In WT mice, PFOS induced changes that were characteristic of PPARα transactivation including regulation of genes associated with lipid metabolism, peroxisome biogenesis, proteasome activation, and inflammation. PPARα-independent changes were indicated in both WT and Null mice by altered expression of genes related to lipid metabolism, inflammation, and xenobiotic metabolism. Such results are similar to studies done with PFOA and are consistent with modest activation of the constitutive androstane receptor (CAR), and possibly PPARγ and/or PPARß/δ. Unique treatment-related effects were also found in Null mice including altered expression of genes associated with ribosome biogenesis, oxidative phosphorylation, and cholesterol biosynthesis. Of interest was up-regulation of Cyp7a1, a gene which is under the control of various transcription regulators. Hence, in addition to its ability to modestly activate PPARα, PFOS induces a variety of PPARα-independent effects as well.

10.
PPAR Res ; 20102010.
Artigo em Inglês | MEDLINE | ID: mdl-20706641

RESUMO

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.

11.
Reprod Toxicol ; 27(3-4): 278-288, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19429403

RESUMO

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.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Caprilatos/toxicidade , Fluorocarbonos/toxicidade , Perfilação da Expressão Gênica , Fígado/metabolismo , Pulmão/metabolismo , Ácidos Alcanossulfônicos/farmacologia , Animais , Caprilatos/farmacologia , Relação Dose-Resposta a Droga , Feminino , Feto/metabolismo , Fluorocarbonos/farmacologia , Exposição Materna , Camundongos , Camundongos Endogâmicos , Análise em Microsséries , Gravidez
12.
Reprod Toxicol ; 27(3-4): 258-265, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18595657

RESUMO

Perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) are members of a family of perfluorinated compounds. Both are environmentally persistent and found in the serum of wildlife and humans. PFOS and PFOA are developmentally toxic in laboratory rodents. Exposure to these chemicals in utero delays development and reduces postnatal survival and growth. Exposure to PFOS on the last 4 days of gestation in the rat is sufficient to reduce neonatal survival. PFOS and PFOA are weak agonists of peroxisome proliferator activated receptor-alpha (PPAR alpha). The reduced postnatal survival of neonatal mice exposed to PFOA was recently shown to depend on expression of PPAR alpha. This study used PPAR alpha knockout (KO) and 129S1/SvlmJ wild type (WT) mice to determine if PPAR alpha expression is required for the developmental toxicity of PFOS. After mating overnight, the next day was designated gestation day (GD) 0. WT females were weighed and dosed orally from GD15 to 18 with 0.5% Tween-20, 4.5, 6.5, 8.5, or 10.5mg PFOS/kg/day. KO females were dosed with 0.5% Tween-20, 8.5 or 10.5mg PFOS/kg/day. Dams and pups were observed daily and pups were weighed on postnatal day (PND) 1 and PND15. Eye opening was recorded from PND12 to 15. Dams and pups were killed on PND15, body and liver weights recorded, and serum collected. PFOS did not affect maternal weight gain or body or liver weights of the dams on PND15. Neonatal survival (PND1-15) was significantly reduced by PFOS in both WT and KO litters at all doses. WT and KO pup birth weight and weight gain from PND1 to 15 were not significantly affected by PFOS exposure. Relative liver weight of WT and KO pups was significantly increased by the 10.5mg/kg dose. Eye opening of PFOS-exposed pups was slightly delayed in WT and KO on PND13 or 14, respectively. Because results in WT and KO were comparable, it is concluded that PFOS-induced neonatal lethality and delayed eye opening are not dependent on activation of PPAR alpha.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Poluentes Ambientais/toxicidade , Fluorocarbonos/toxicidade , PPAR alfa/metabolismo , Ácidos Alcanossulfônicos/agonistas , Animais , Peso ao Nascer , Relação Dose-Resposta a Droga , Poluentes Ambientais/agonistas , Olho/efeitos dos fármacos , Olho/crescimento & desenvolvimento , Feminino , Fluorocarbonos/agonistas , Fígado/efeitos dos fármacos , Fígado/crescimento & desenvolvimento , Masculino , Exposição Materna , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Tamanho do Órgão/efeitos dos fármacos , Fatores Sexuais , Análise de Sobrevida
13.
Toxicol Sci ; 105(1): 173-81, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18511431

RESUMO

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.


Assuntos
Feto/efeitos dos fármacos , Fluorocarbonos/toxicidade , Anormalidades Induzidas por Medicamentos , Animais , Peso Corporal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Fluorocarbonos/sangue , Expressão Gênica/efeitos dos fármacos , Crescimento/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Tamanho do Órgão/efeitos dos fármacos , Gravidez
14.
Toxicol Pathol ; 36(4): 592-607, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18467677

RESUMO

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.


Assuntos
Caprilatos/toxicidade , Poluentes Ambientais/toxicidade , Fluorocarbonos/toxicidade , Perfilação da Expressão Gênica , Expressão Gênica/efeitos dos fármacos , Fígado/efeitos dos fármacos , PPAR alfa/metabolismo , Animais , Caprilatos/farmacocinética , Poluentes Ambientais/farmacocinética , Fluorocarbonos/farmacocinética , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , PPAR alfa/genética , Pirimidinas/toxicidade , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Toxicol Pathol ; 36(4): 632-9, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18467680

RESUMO

Perfluorooctanoic acid (PFOA) is a chemical used in the production of fluoropolymers. Its persistence in the environment and presence in humans and wildlife has raised health concerns. Liver tumor induction by PFOA is thought to be mediated in rodents by PPAR-alpha. A recent US EPA scientific advisory board questioned the contribution of PPAR-alpha in PFOA-induced liver tumors. Liver response in CD-1, SV/129 wild-type (WT), and PPAR-alpha knockout (KO) SV/129 mice was evaluated after seven daily treatments of PFOA-NH4(+) (1, 3, or 10 mg/kg, p.o.) or the prototype PPARalpha-agonist Wyeth 14,643 (WY, 50 mg/kg). Livers were examined by light and electron microscopy. Proliferation was quantified after PCNA immunostaining. PFOA treatment induced a dose-dependent increase in hepatocyte hypertrophy and labeling index (LI) similar to WY in WT mice. Ultrastructural alterations of peroxisome proliferation were similar between WY-treated and 10 mg/kg PFOA-treated WT mice. KO mice had a dose-dependent increase in hepatocyte vacuolation but increased LI only at 10 mg PFOA/kg. WY-treated KO mice were not different from KO control. These data suggest that PPAR-alpha is required for WY- and PFOA-induced cellular alterations in WT mouse liver. Hepatic enlargement observed in KO mice may be due to an accumulation of cytoplasmic vacuoles that contain PFOA.


Assuntos
Caprilatos/toxicidade , Poluentes Ambientais/toxicidade , Fluorocarbonos/toxicidade , Fígado/efeitos dos fármacos , PPAR alfa/fisiologia , Pirimidinas/toxicidade , Animais , Caprilatos/sangue , Caprilatos/farmacocinética , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Poluentes Ambientais/sangue , Poluentes Ambientais/farmacocinética , Fluorocarbonos/sangue , Fluorocarbonos/farmacocinética , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/ultraestrutura , Fígado/metabolismo , Fígado/ultraestrutura , Camundongos , Camundongos Knockout , Tamanho do Órgão/efeitos dos fármacos , PPAR alfa/agonistas , PPAR alfa/genética , Pirimidinas/sangue , Pirimidinas/farmacocinética
16.
Toxicol Sci ; 98(2): 571-81, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17488742

RESUMO

Perfluorooctanoic acid (PFOA) is a member of a family of perfluorinated chemicals that have a variety of applications. PFOA persists in the environment and is found in wildlife and humans. In mice, PFOA is developmentally toxic producing mortality, delayed eye opening, growth deficits, and altered pubertal maturation. PFOA activates peroxisome proliferators-activated receptor-alpha (PPARalpha), a pathway critical to the mode of induction of liver tumors in rodents. The present study uses 129S1/SvlmJ wild-type (WT) and PPARalpha knockout (KO) mice to determine if PPARalpha mediates PFOA-induced developmental toxicity. Pregnant mice were dosed orally from gestation days 1-17 with water or 0.1, 0.3, 0.6, 1, 3, 5, 10, or 20 mg PFOA/kg. PFOA did not affect maternal weight, embryonic implantation, number, or weight of pups at birth. At 5 mg/kg, the incidence of full litter resorptions increased in both WT and KO mice. In WT, but not KO, neonatal survival was reduced (0.6 mg/kg) and eye opening was delayed (1 mg/kg). There was a trend across dose for reduced pup weight (WT and KO) on several postnatal days (PND), but only WT exposed to 1 mg/kg were significantly different from control (PND7-10 and 22). Maternal factors (e.g., background genetics) did not contribute to differences in postnatal mortality, as PFOA induced postnatal mortality in heterozygous pups born to WT or KO dams. In conclusion, early pregnancy loss was independent of PPARalpha expression. Delayed eye opening and deficits in postnatal weight gain appeared to depend on PPARalpha expression, although other mechanisms may contribute. PPARalpha was required for PFOA-induced postnatal lethality and expression of one copy of the gene was sufficient to mediate this effect.


Assuntos
Caprilatos/toxicidade , Perda do Embrião/induzido quimicamente , Fluorocarbonos/toxicidade , PPAR alfa/genética , Animais , Caprilatos/sangue , Caprilatos/farmacocinética , Perda do Embrião/sangue , Perda do Embrião/genética , Olho/efeitos dos fármacos , Olho/crescimento & desenvolvimento , Feminino , Fluorocarbonos/sangue , Fluorocarbonos/farmacocinética , Fígado/efeitos dos fármacos , Fígado/crescimento & desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Tamanho do Órgão/efeitos dos fármacos , PPAR alfa/deficiência , Gravidez
17.
Toxicol Sci ; 95(2): 462-73, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17098816

RESUMO

Perfluorooctanoic acid (PFOA) is a persistent pollutant and is detectable in human serum (5 ng/ml in the general population of the Unites States). PFOA is used in the production of fluoropolymers which have applications in the manufacture of a variety of industrial and commercial products (e.g., textiles, house wares, electronics). PFOA is developmentally toxic and in mice affects growth, development, and viability of offspring. This study segregates the contributions of gestational and lactational exposures and considers the impact of restricting exposure to specific gestational periods. Pregnant CD-1 mice were dosed on gestation days (GD) 1-17 with 0, 3, or 5 mg PFOA/kg body weight, and pups were fostered at birth to give seven treatment groups: unexposed controls, pups exposed in utero (3U and 5U), lactationally (3L and 5L), or in utero + lactationally (3U + L and 5U + L). In the restricted exposure (RE) study, pregnant mice received 5 mg PFOA/kg from GD7-17, 10-17, 13-17, or 15-17 or 20 mg on GD15-17. In all PFOA-treated groups, dam weight gain, number of implantations, and live litter size were not adversely affected and relative liver weight increased. Treatment with 5 mg/kg on GD1-17 increased the incidence of whole litter loss and pups in surviving litters had reduced birth weights, but effects on pup survival from birth to weaning were only affected in 5U + L litters. In utero exposure (5U), in the absence of lactational exposure, was sufficient to produce postnatal body weight deficits and developmental delay in the pups. In the RE study, birth weight and survival were reduced by 20 mg/kg on GD15-17. Birth weight was also reduced by 5 mg/kg on GD7-17 and 10-17. Although all PFOA-exposed pups had deficits in postnatal weight gain, only those exposed on GD7-17 and 10-17 also showed developmental delay in eye opening and hair growth. In conclusion, the postnatal developmental effects of PFOA are due to gestational exposure. Exposure earlier in gestation produced stronger responses, but further study is needed to determine if this is a function of higher total dose or if there is a developmentally sensitive period.


Assuntos
Peso Corporal/efeitos dos fármacos , Caprilatos/toxicidade , Poluentes Ambientais/toxicidade , Fluorocarbonos/toxicidade , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Animais , Peso ao Nascer/efeitos dos fármacos , Caprilatos/sangue , Poluentes Ambientais/sangue , Feminino , Fluorocarbonos/sangue , Idade Gestacional , Lactação , Tamanho da Ninhada de Vivíparos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos , Tamanho do Órgão/efeitos dos fármacos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/sangue
18.
Neurotoxicol Teratol ; 26(3): 397-406, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15113601

RESUMO

In vitro techniques are used increasingly to screen for and characterize neurotoxicants. In many cases, chemical-induced injury to developing neurons has been examined in vitro by assessing morphological changes in differentiation and neurite growth. This research evaluated the use of proteins associated with axonal growth and synaptogenesis as surrogates for morphological measurement of neuronal differentiation. PC12 cells, which differentiate upon nerve growth factor (NGF) stimulation, were used as the in vitro model. NGF-induced (50 ng/ml) differentiation (cells with at least one neurite with a length equal to the cell body diameter) and neurite growth (length of longest neurite) were determined using light microscopy and computer-based quantitative image analysis. PC12 cell differentiation and neurite growth reached a plateau after 6 days in culture. Expression of the axonal growth associated protein 43 (GAP-43) and the synaptic protein synapsin I were assessed simultaneously by Western blot during cell differentiation. Expression of GAP-43 was low on Culture Day 0 and increased progressively to maximum levels on Culture Day 5. Likewise, synapsin I expression increased slowly on Days 0-4, and then rapidly on Days 5-7 of culture. Pharmacologic inhibitors of NGF-induced signaling were used to test the sensitivity of the proteins to chemical disruption of differentiation. The MAP kinase inhibitor, U0126 (5-30 microM) and the PKC inhibitor, bisindolylmaleimide I (Bis I; 1.25-5 microM) inhibited differentiation and neurite outgrowth in a concentration-dependent manner. U0126 and Bis I significantly decreased GAP-43, but not synapsin I expression. Interestingly, the PI-PLC inhibitor edelfosine (ET-18; 5-30 microM) stimulated differentiation at early times of exposure followed by a significant decrease in neurite length at later time points. However, ET-18 did not alter the expression of GAP-43 or synapsin I. These data suggest that GAP-43 may be a useful indicator of the status of PC12 cell differentiation.


Assuntos
Diferenciação Celular/fisiologia , Neuritos/fisiologia , Neuroquímica , Células PC12/citologia , Animais , Western Blotting/métodos , Diferenciação Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Proteína GAP-43/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Processamento de Imagem Assistida por Computador/métodos , Fator de Crescimento Neural/farmacologia , Neuritos/efeitos dos fármacos , Ratos , Sinapsinas/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...