Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Birth Defects Res ; 112(1): 19-39, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31471948

ABSTRACT

Cleft palate has been linked to both genetic and environmental factors that perturb key events during palatal morphogenesis. As a developmental outcome, it presents a challenging, mechanistically complex endpoint for predictive modeling. A data set of 500 chemicals evaluated for their ability to induce cleft palate in animal prenatal developmental studies was compiled from Toxicity Reference Database and the biomedical literature, which included 63 cleft palate active and 437 inactive chemicals. To characterize the potential molecular targets for chemical-induced cleft palate, we mined the ToxCast high-throughput screening database for patterns and linkages in bioactivity profiles and chemical structural descriptors. ToxCast assay results were filtered for cytotoxicity and grouped by target gene activity to produce a "gene score." Following unsuccessful attempts to derive a global prediction model using structural and gene score descriptors, hierarchical clustering was applied to the set of 63 cleft palate positives to extract local structure-bioactivity clusters for follow-up study. Patterns of enrichment were confirmed on the complete data set, that is, including cleft palate inactives, and putative molecular initiating events identified. The clusters corresponded to ToxCast assays for cytochrome P450s, G-protein coupled receptors, retinoic acid receptors, the glucocorticoid receptor, and tyrosine kinases/phosphatases. These patterns and linkages were organized into preliminary decision trees and the resulting inferences were mapped to a putative adverse outcome pathway framework for cleft palate supported by literature evidence of current mechanistic understanding. This general data-driven approach offers a promising avenue for mining chemical-bioassay drivers of complex developmental endpoints where data are often limited.


Subject(s)
Cleft Palate/etiology , Small Molecule Libraries/analysis , Toxicity Tests/methods , Cluster Analysis , Databases, Factual , Female , Follow-Up Studies , High-Throughput Screening Assays/methods , Humans , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Risk Assessment
2.
Methods Mol Biol ; 1965: 93-105, 2019.
Article in English | MEDLINE | ID: mdl-31069670

ABSTRACT

The morphogenesis of the secondary palate provides an interesting model for many of the processes involved in embryonic development. A number of in vitro models have been used to study craniofacial development, including whole embryo culture, palatal mesenchymal and micromass cell cultures, and Trowell-like palatal cultures in which dissected palates are cultured individually or as pairs in contact on a support above medium. This chapter presents a detailed protocol for the culture of maxillary midfacial tissues, including the palatal shelves, in suspension culture. This method involves isolation of the midfacial tissues (maxillary arch and palatal shelves) and suspension of the tissues in medium in flasks. On a rocker in an incubator, the palatal shelves elevate, grow, make contact, and fuse in a time span analogous to that occurring in the intact embryo in utero.


Subject(s)
Palate/cytology , Tissue Culture Techniques/instrumentation , Animals , Cell Proliferation , Cells, Cultured , Incubators , Mice , Models, Biological
3.
Toxicol Sci ; 166(2): 394-408, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30496568

ABSTRACT

Embryologic development involves cell differentiation and organization events that are unique to each tissue and organ and are susceptible to developmental toxicants. Animal models are the gold standard for identifying putative teratogens, but the limited throughput of developmental toxicological studies in animals coupled with the limited concordance between animal and human teratogenicity motivates a different approach. In vitro organoid models can mimic the three-dimensional (3D) morphogenesis of developing tissues and can thus be useful tools for studying developmental toxicology. Common themes during development like the involvement of epithelial-mesenchymal transition and tissue fusion present an opportunity to develop in vitro organoid models that capture key morphogenesis events that occur in the embryo. We previously described organoids composed of human stem and progenitor cells that recapitulated the cellular features of palate fusion, and here we further characterized the model by examining pharmacological inhibitors targeting known palatogenesis and epithelial morphogenesis pathways as well as 12 cleft palate teratogens identified from rodent models. Organoid survival was dependent on signaling through EGF, IGF, HGF, and FGF pathways, and organoid fusion was disrupted by inhibition of BMP signaling. We observed concordance between the effects of EGF, FGF, and BMP inhibitors on organoid fusion and epithelial cell migration in vitro, suggesting that organoid fusion is dependent on epithelial morphogenesis. Three of the 12 putative cleft palate teratogens studied here (theophylline, triamcinolone, and valproic acid) significantly disrupted in vitro organoid fusion, while tributyltin chloride and all-trans retinoic acid were cytotoxic to fusing organoids. The study herein demonstrates the utility of the in vitro fusion assay for identifying chemicals that disrupt human organoid morphogenesis in a scalable format amenable to toxicology screening.


Subject(s)
Morphogenesis/drug effects , Organ Culture Techniques/methods , Organoids/drug effects , Palate/drug effects , Palate/embryology , Teratogens/pharmacology , Aminopyridines/pharmacology , Anilides/pharmacology , Benzazepines/pharmacology , Benzimidazoles/pharmacology , Cell Movement/drug effects , Cell Survival/drug effects , Epidermal Cells/drug effects , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Indoles/pharmacology , Keratin-17/metabolism , Mesenchymal Stem Cells/drug effects , Organoids/metabolism , Phenols/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyridones/pharmacology , Spheroids, Cellular , Staurosporine/pharmacology , Stem Cells/drug effects , Sulfones/pharmacology , Vimentin/metabolism
4.
Birth Defects Res ; 110(17): 1322-1334, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30347137

ABSTRACT

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.


Subject(s)
Bone Development/physiology , Cleft Palate/embryology , Epidermal Growth Factor/metabolism , Epithelial Cells/cytology , Mesenchymal Stem Cells/cytology , Palate/embryology , Cell Proliferation/physiology , Cells, Cultured , Humans , Osteogenesis/physiology , Spheroids, Cellular/cytology , Umbilical Veins/cytology
5.
PLoS One ; 12(9): e0184155, 2017.
Article in English | MEDLINE | ID: mdl-28898253

ABSTRACT

Epithelial-mesenchymal interactions drive embryonic fusion events during development, and perturbations of these interactions can result in birth defects. Cleft palate and neural tube defects can result from genetic defects or environmental exposures during development, yet very little is known about the effect of chemical exposures on fusion events during human development because of a lack of relevant and robust human in vitro assays of developmental fusion behavior. Given the etiology and prevalence of cleft palate and the relatively simple architecture and composition of the embryonic palate, we sought to develop a three-dimensional culture system that mimics the embryonic palate and could be used to study fusion behavior in vitro using human cells. We engineered size-controlled human Wharton's Jelly stromal cell (HWJSC) spheroids and established that 7 days of culture in osteogenesis differentiation medium was sufficient to promote an osteogenic phenotype consistent with embryonic palatal mesenchyme. HWJSC spheroids supported the attachment of human epidermal keratinocyte progenitor cells (HPEKp) on the outer spheroid surface likely through deposition of collagens I and IV, fibronectin, and laminin by mesenchymal spheroids. HWJSC spheroids coated in HPEKp cells exhibited fusion behavior in culture, as indicated by the removal of epithelial cells from the seams between spheroids, that was dependent on epidermal growth factor signaling and fibroblast growth factor signaling in agreement with palate fusion literature. The method described here may broadly apply to the generation of three-dimensional epithelial-mesenchymal co-cultures to study developmental fusion events in a format that is amenable to predictive toxicology applications.


Subject(s)
Bioengineering , Organ Culture Techniques , Palate/embryology , Spheroids, Cellular , Alkaline Phosphatase/metabolism , Bioengineering/methods , Cell Differentiation/genetics , Cluster Analysis , Computational Biology/methods , Extracellular Matrix Proteins , Gene Expression Profiling , Gene Ontology , Humans , In Vitro Techniques , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics , Palate/metabolism , Time Factors , Transcriptome
6.
Toxicology ; 382: 93-107, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28285100

ABSTRACT

Crosstalk between epithelial and stromal cells drives the morphogenesis of ectodermal organs during development and promotes normal mature adult epithelial tissue homeostasis. Epithelial-stromal interactions (ESIs) have historically been examined using mammalian models and ex vivo tissue recombination. Although these approaches have elucidated signaling mechanisms underlying embryonic morphogenesis processes and adult mammalian epithelial tissue function, they are limited by the availability of tissue, low throughput, and human developmental or physiological relevance. In this review, we describe how bioengineered ESIs, using either human stem cells or co-cultures of human primary epithelial and stromal cells, have enabled the development of human in vitro epithelial tissue models that recapitulate the architecture, phenotype, and function of adult human epithelial tissues. We discuss how the strategies used to engineer mature epithelial tissue models in vitro could be extrapolated to instruct the design of organotypic culture models that can recapitulate the structure of embryonic ectodermal tissues and enable the in vitro assessment of events critical to organ/tissue morphogenesis. Given the importance of ESIs towards normal epithelial tissue development and function, such models present a unique opportunity for toxicological screening assays to incorporate ESIs to assess the impact of chemicals on mature and developing epidermal tissues.


Subject(s)
Epithelial Cells , Stromal Cells , Animals , Bioengineering , Coculture Techniques , Humans , Toxicity Tests
7.
Toxicology ; 378: 37-52, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28049043

ABSTRACT

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.


Subject(s)
Alkanesulfonic Acids/toxicity , Environmental Pollutants/toxicity , Fatty Liver/chemically induced , Fatty Liver/genetics , Fluorocarbons/toxicity , Lipid Metabolism/genetics , Animals , Cell Line, Tumor , DNA/metabolism , Fatty Acids/metabolism , Fatty Liver/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Homeostasis/drug effects , Humans , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Knockout , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Mitochondria, Liver/physiology , PPAR alpha/genetics , Palmitates/metabolism , Rats, Sprague-Dawley , Triglycerides/metabolism
8.
Reprod Toxicol ; 65: 321-358, 2016 10.
Article in English | MEDLINE | ID: mdl-27575429

ABSTRACT

The 2011 EPA trichloroethylene (TCE) IRIS assessment, used developmental cardiac defects from a controversial drinking water study in rats (Johnson et al. [51]), along with several other studies/endpoints to derive reference values. An updated literature search of TCE-related developmental cardiac defects was conducted. Study quality, strengths, and limitations were assessed. A putative adverse outcome pathway (AOP) construct was developed to explore key events for the most commonly observed cardiac dysmorphologies, particularly those involved with epithelial-mesenchymal transition (EMT) of endothelial origin (EndMT); several candidate pathways were identified. A hypothesis-driven weight-of-evidence analysis of epidemiological, toxicological, in vitro, in ovo, and mechanistic/AOP data concluded that TCE has the potential to cause cardiac defects in humans when exposure occurs at sufficient doses during a sensitive window of fetal development. The study by Johnson et al. [51] was reaffirmed as suitable for hazard characterization and reference value derivation, though acknowledging study limitations and uncertainties.


Subject(s)
Environmental Exposure/adverse effects , Environmental Pollutants/toxicity , Heart/drug effects , Prenatal Exposure Delayed Effects , Solvents/toxicity , Trichloroethylene/toxicity , Animals , Dose-Response Relationship, Drug , Epithelial-Mesenchymal Transition , Female , Heart/embryology , Humans , Pregnancy
9.
Mol Cell Endocrinol ; 400: 90-101, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25448844

ABSTRACT

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.


Subject(s)
Adipocytes/drug effects , Environmental Pollutants/pharmacology , Gene Expression Regulation/drug effects , Lipid Metabolism/drug effects , Stearoyl-CoA Desaturase/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Alkanesulfonic Acids/pharmacology , Animals , Caprylates/pharmacology , Cell Differentiation/drug effects , Cell Size , Fatty Acids , Fluorocarbons/pharmacology , Gene Expression Profiling , Mice , PPAR alpha/agonists , PPAR alpha/genetics , PPAR alpha/metabolism , PPAR gamma/agonists , PPAR gamma/genetics , PPAR gamma/metabolism , Pyrimidines/pharmacology , Rosiglitazone , Signal Transduction , Stearoyl-CoA Desaturase/genetics , Thiazolidinediones/pharmacology
10.
Toxicology ; 316: 43-54, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24374136

ABSTRACT

Perfluoroalkyl acids (PFAAs) are found globally in the environment, detected in humans and wildlife, and are typically present as mixtures of PFAA congeners. Mechanistic studies have found that responses to PFAAs are mediated in part by PPARα. Our previous studies showed that individual PFAAs activate PPARα transfected into COS-1 cells. The goal of the current study was to determine if binary combinations of perfluorooctanoic acid (PFOA) and another PFAA act in an additive fashion to activate PPARα in the mouse one-hybrid in vitro model. COS-1 cells were transiently transfected with mouse PPARα luciferase reporter construct and exposed to either vehicle control (0.1% DMSO or water), PPARα agonist (WY14643, 10 µM), PFOA at 1-128µM, perfluorononanoic acid (PFNA) at 1-128 µM, perfluorohexanoic acid (PFHxA) at 8-1024 µM, perfluorooctane sulfonate (PFOS) at 4-384 µM or perfluorohexane sulfonate (PFHxS) at 8-2048 µM to generate sigmoidal concentration-response curves. In addition, cells were exposed to binary combinations of PFOA+either PFNA, PFHxA, PFOS or PFHxS in an 8×8 factorial design. The concentration-response data for individual chemicals were fit to sigmoidal curves and analyzed with nonlinear regression to generate EC50s and Hillslopes, which were used in response-addition and concentration-addition models to calculate predicted responses for mixtures in the same plate. All PFOA+PFAA combinations produced concentration-response curves that were closely aligned with the predicted curves for both response addition and concentration addition at low concentrations. However, at higher concentrations of all chemicals, the observed response curves deviated from the predicted models of additivity. We conclude that binary combinations of PFAAs behave additively at the lower concentration ranges in activating PPARα in this in vitro system.


Subject(s)
Caprylates/toxicity , Environmental Pollutants/toxicity , Fluorocarbons/toxicity , PPAR alpha/drug effects , Alkanesulfonic Acids/administration & dosage , Alkanesulfonic Acids/chemistry , Alkanesulfonic Acids/toxicity , Animals , COS Cells , Caprylates/administration & dosage , Caprylates/chemistry , Chlorocebus aethiops , Dose-Response Relationship, Drug , Environmental Pollutants/administration & dosage , Environmental Pollutants/chemistry , Fluorocarbons/administration & dosage , Fluorocarbons/chemistry , Mice , PPAR alpha/metabolism , Pyrimidines/pharmacology , Regression Analysis , Transfection
11.
Toxicology ; 308: 129-37, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23567314

ABSTRACT

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.


Subject(s)
Alkanesulfonic Acids/chemistry , Alkanesulfonic Acids/toxicity , Fluorocarbons/chemistry , Fluorocarbons/toxicity , Hepatocytes/drug effects , Animals , Drug Evaluation, Preclinical/methods , Humans , Mice , Primary Cell Culture
12.
Toxicology ; 306: 169-75, 2013 Apr 05.
Article in English | MEDLINE | ID: mdl-23470359

ABSTRACT

This study is a follow-up to a paper by Carr et al. that determined a design structure to optimally test for departures from additivity in a fixed ratio mixture of four perfluoroalkyl acids (PFAAs) using an in vitro transiently-transfected COS-1 PPARα reporter model with a mixing ratio that is based on average serum levels in NHANES subjects. Availability of information regarding potential for additivity of PFAAs in mixtures is critically important for risk assessors who are concerned with the ability of the compounds to affect human health and impact ecological systems. It is clear that exposures are not to single compounds, but to mixtures of the PFAAs. This paper presents the results from the data collected using the design from Carr et al. along with subsequent analyses that were performed to classify the relationships among mixtures of PFAAs. A non-linear logistic additivity model was employed to predict relative luciferase units (RLU), an indicator of PPARα activation. The results indicated a less than additive relationship among the four PFAAs. To determine if the possible "antagonism" is from the competition among or between carboxylates and sulfonates, four different binary mixtures were also studied. There was a less than additive relationship in all four binary mixtures. These findings are generally similar to two other reports of interfering interactions between PFAAs in mixtures. The most conservative interpretation for our data would be an assumption of additivity (and lack of a greater than additive interaction), with a potential for antagonistic interactions.


Subject(s)
Alkanesulfonates/chemistry , Alkanesulfonates/toxicity , Carboxylic Acids/chemistry , Carboxylic Acids/toxicity , Models, Statistical , Alkanesulfonates/metabolism , Animals , COS Cells , Carboxylic Acids/metabolism , Chlorocebus aethiops , Complex Mixtures/chemistry , Complex Mixtures/metabolism , Complex Mixtures/toxicity , PPAR alpha/metabolism
13.
Reprod Toxicol ; 33(4): 546-551, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22107727

ABSTRACT

Perfluorinated alkyl acids (PFAAs) are manufactured surfactants found globally in the environment and in tissues of humans and wildlife. Several PFAAs adversely affect rodents and activation of PPARα is thought to be their mode of action. Our previous study demonstrated that some PFAAs activate mouse and human PPARα in transiently transfected COS-1 cells. Here, we test more PFAAs for PPARα activation in the same system. Cells were transfected with either mouse or human PPARα-luciferase reporter plasmid, exposed the next day to either vehicle, PPARα agonist (WY14643), perfluoropentanoic acid (C5), perfluoroheptanoic acid (C7), perfluorooctanoic acid (C8), perfluoroundecanoic acid (C11), or perfluorododecanoic acid (C12) at concentrations from 0.5µM to 100µM, and luminescence was measured after 24h. C8 induced the highest activity for human PPARα, followed by C7, C5, and C11. C12 had little activity. C8 induced the highest activity for mouse PPARα, followed by C11, C7, C12 and C5. The two studies together found increasing activity of PPARα with increasing chain length of the PFAA up to perfluorononanoic acid (C9) and lower activity with longer chain PFAAs with both mouse and human PPARα.


Subject(s)
Environmental Pollutants/chemistry , Environmental Pollutants/toxicity , Fluorocarbons/chemistry , Fluorocarbons/toxicity , PPAR alpha/metabolism , Animals , COS Cells , Cell Culture Techniques , Cell Survival/drug effects , Chlorocebus aethiops , Dose-Response Relationship, Drug , Humans , Linear Models , Mice , No-Observed-Adverse-Effect Level , PPAR alpha/genetics , Plasmids , Species Specificity , Structure-Activity Relationship , Transfection
14.
Reprod Toxicol ; 33(4): 491-505, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22154759

ABSTRACT

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.


Subject(s)
Caprylates/toxicity , Environmental Pollutants/toxicity , Fetal Development , Fluorocarbons/toxicity , Gene Expression Regulation, Developmental/drug effects , Peroxisome Proliferator-Activated Receptors/genetics , Prenatal Exposure Delayed Effects , Aging/genetics , Aging/metabolism , Animals , Animals, Newborn , Blotting, Western , Caprylates/blood , Caprylates/pharmacokinetics , Environmental Pollutants/blood , Environmental Pollutants/pharmacokinetics , Female , Fetal Development/drug effects , Fetal Development/genetics , Fluorocarbons/blood , Fluorocarbons/pharmacokinetics , Gestational Age , Male , Mice , Mice, Inbred Strains , Organ Specificity , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/metabolism , Real-Time Polymerase Chain Reaction
15.
PPAR Res ; 20102010.
Article in English | MEDLINE | ID: mdl-20936102

ABSTRACT

Perfluorononanoic acid (PFNA) is one of the perfluoroalkyl acids found in the environment and in tissues of humans and wildlife. Prenatal exposure to PFNA negatively impacts survival and development of mice and activates the mouse and human peroxisome proliferator-activated receptor-alpha (PPARα). In the current study, we used PPARα knockout (KO) and 129S1/SvlmJ wild-type (WT) mice to investigate the role of PPARα in mediating PFNA-induced in vivo effects. Pregnant KO and WT mice were dosed orally with water (vehicle control: 10 ml/kg), 0.83, 1.1, 1.5, or 2 mg/kg PFNA on gestational days (GDs) 1-18 (day of sperm plug = GD 0). Maternal weight gain, implantation, litter size, and pup weight at birth were unaffected in either strain. PFNA exposure reduced the number of live pups at birth and survival of offspring to weaning in the 1.1 and 2 mg/kg groups in WT. Eye opening was delayed (mean delay 2.1 days) and pup weight at weaning was reduced in WT pups at 2 mg/kg. These developmental endpoints were not affected in the KO. Relative liver weight was increased in a dose-dependent manner in dams and pups of the WT strain at all dose levels but only slightly increased in the highest dose group in the KO strain. In summary, PFNA altered liver weight of dams and pups, pup survival, body weight, and development in the WT, while only inducing a slight increase in relative liver weight of dams and pups at 2 mg/kg in KO mice. These results suggest that PPARα is an essential mediator of PFNA-induced developmental toxicity in the mouse.

16.
PPAR Res ; 20102010.
Article in English | MEDLINE | ID: mdl-20936131

ABSTRACT

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.

17.
PPAR Res ; 20102010.
Article in English | MEDLINE | ID: mdl-20706641

ABSTRACT

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.

18.
Birth Defects Res B Dev Reprod Toxicol ; 89(4): 266-74, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20602452

ABSTRACT

Dates of special, historical significance, such as the 50th anniversary of the founding of the Teratology Society, prompt a desire to pause and look back and contemplate where we began, how far we have come, and consider the future for our scientific endeavors. The study of the etiology of cleft palate extends many years into the past and was a subject of interest to many of the founding members of the Teratology Society. This research area was intensively pursued and spawned a vast portfolio of published research. This article will look back at the state of the science around the time of the founding of the Teratology Society, in the 1950s and 1960s, and track the emergence and pursuit of an interest in an etiology for cleft palate involving failure of palatal fusion. Studies of medial epithelial cell fate and induction of cleft palate by interference with adhesion or fusion span the period from the 1960s to the present time. Teratology Society members have been and continue to be key players in cleft palate research. In this retrospective article, seminal research published by Teratology Society members will serve as a platform to launch the discussion of the emergence of our current understanding of medial epithelial cell differentiation and fate and the potential for these processes to be targets of teratogenic action.


Subject(s)
Cleft Palate/embryology , Cleft Palate/etiology , Palate/embryology , Animals , Cell Differentiation , Cleft Palate/genetics , Disease Models, Animal , Epithelial Cells/metabolism , History, 20th Century , History, 21st Century , Mice , Teratogens/pharmacology
20.
Reprod Toxicol ; 27(3-4): 246-257, 2009 Jun.
Article in English | MEDLINE | ID: mdl-18996469

ABSTRACT

The peroxisome proliferator-activated receptors (PPAR) belong to the nuclear hormone receptor superfamily and there are three primary subtypes, PPARalpha, beta, and gamma. These receptors regulate important physiological processes that impact lipid homeostasis, inflammation, adipogenesis, reproduction, wound healing, and carcinogenesis. These nuclear receptors have important roles in reproduction and development and their expression may influence the responses of an embryo exposed to PPAR agonists. PPARs are relevant to the study of the biological effects of the perfluorinated alkyl acids as these compounds, including perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS), activate PPARalpha. Exposure of the rodent to PFOA or PFOS during gestation results in neonatal deaths, developmental delay and growth deficits. Studies in PPARalpha knockout mice demonstrate that the developmental effects of PFOA, but not PFOS, depend on expression of PPARalpha. This review provides an overview of PPARalpha, beta, and gamma protein and mRNA expression during mouse, rat, and human development. The review presents the results from many published studies and the information is organized by organ system and collated to show patterns of expression at comparable developmental stages for human, mouse, and rat. The features of the PPAR nuclear receptor family are introduced and what is known or inferred about their roles in development is discussed relative to insights from genetically modified mice and studies in the adult.


Subject(s)
Human Development , PPAR alpha/metabolism , PPAR gamma/metabolism , PPAR-beta/metabolism , Rodentia/embryology , Rodentia/growth & development , Animals , Gene Expression Regulation, Developmental , Humans , Mice , Mice, Knockout , PPAR alpha/genetics , PPAR gamma/genetics , PPAR-beta/genetics , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...