Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Environ Int ; 167: 107385, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35952468

RESUMO

BACKGROUND: Environmental health research has recently undergone a dramatic shift, with ongoing technological advancements allowing for broader coverage of exposure and molecular biology signatures. Approaches to integrate such measures are still needed to increase understanding between systems-level exposure and biology. OBJECTIVES: We address this gap by evaluating placental tissues to identify novel chemical-biological interactions associated with preeclampsia. This study tests the hypothesis that understudied chemicals are present in the human placenta and associated with preeclampsia-relevant disruptions, including overall case status (preeclamptic vs. normotensive patients) and underlying transcriptomic/epigenomic signatures. METHODS: A non-targeted analysis based on high-resolution mass spectrometry was used to analyze placental tissues from a cohort of 35 patients with preeclampsia (n = 18) and normotensive (n = 17) pregnancies. Molecular feature data were prioritized for confirmation based on association with preeclampsia case status and confidence of chemical identification. All molecular features were evaluated for relationships to mRNA, microRNA, and CpG methylation (i.e., multi-omic) signature alterations involved in preeclampsia. RESULTS: A total of 183 molecular features were identified with significantly differentiated abundance in placental extracts of preeclamptic patients; these features clustered into distinct chemical groupings using unsupervised methods. Of these features, 53 were identified (mapping to 40 distinct chemicals) using chemical standards, fragmentation spectra, and chemical metadata. In general, human metabolites had the largest feature intensities and strongest associations with preeclampsia-relevant multi-omic changes. Exogenous drugs were second most abundant and had fewer associations with multi-omic changes. Other exogenous chemicals (non-drugs) were least abundant and had the fewest associations with multi-omic changes. CONCLUSIONS: These global data trends suggest that human metabolites are heavily intertwined with biological processes involved in preeclampsia etiology, while exogenous chemicals may still impact select transcriptomic/epigenomic processes. This study serves as a demonstration of merging systems exposures with systems biology to better understand chemical-disease relationships.


Assuntos
Pré-Eclâmpsia , Estudos de Coortes , Epigenômica , Feminino , Humanos , Placenta/metabolismo , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/metabolismo , Gravidez , Transcriptoma
2.
J Biochem Mol Toxicol ; 36(7): e23068, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35403300

RESUMO

Since the discovery of calbindin release into the urine during renal injury, there has been growing interest in the utility of this protein as a biomarker of nephrotoxicity. However, little is known about the intrarenal regulation of the release and expression of this calcium-regulating protein during kidney injury. We sought to characterize the time-dependent expression and excretion of the protein calbindin in the distal tubule in comparison to kidney injury molecule-1 (Kim-1), a protein in the proximal tubule, in mice treated with cisplatin. Urine, blood, and kidneys were collected from male C57BL/6 mice treated with vehicle or cisplatin (20 mg/kg ip). Urinary concentrations of calbindin and Kim-1 were elevated by 11.6-fold and 2.5-fold, respectively, within 2 days after cisplatin. Circulating creatinine and blood urea nitrogen levels increased in cisplatin-treated mice by 3 days, confirming the development of acute kidney injury. Time-dependent decreases in intrarenal calbindin protein were observed on Days 3 and 4 and a 200-fold upregulation of calbindin (CALB1) and KIM-1 messenger RNAs (mRNAs) was observed on Day 3. These data suggest that early loss of calbindin protein into the urine along with declines in renal calbindin levels initiates a compensatory induction of mRNA expression at later time points (Days 3 and 4). Understanding the regulation of calbindin during cisplatin nephrotoxicity further enhances its utility as a potential urinary biomarker of kidney damage. The results of the current study support the combined use of a proximal (Kim-1) and distal tubule (calbindin) marker to phenotype acute kidney injury secondary to cisplatin administration.


Assuntos
Injúria Renal Aguda , Antineoplásicos , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/metabolismo , Animais , Antineoplásicos/efeitos adversos , Biomarcadores/metabolismo , Calbindinas/metabolismo , Cisplatino/toxicidade , Rim/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
3.
Toxicol Appl Pharmacol ; 409: 115305, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33129825

RESUMO

Exposure to inorganic arsenic (iAs) is a significant public health concern with individuals around the globe exposed to harmful levels through contaminated drinking water. Exposure to iAs during pregnancy is of particular concern and has been associated with pregnancy complications and adverse child health later in life. Effects of in utero exposure may be mediated through alterations in key signaling pathways in the placenta that regulate fetal growth and development. A pathway of interest is the glucocorticoid receptor (GR)- signaling pathway, which is known to regulate fetal and placental development. While prior research has shown that iAs alters GR-associated gene expression in trophoblasts, the mechanisms that underlie these perturbations remain unknown. In the present study, we set out to elucidate the molecular mechanisms that underpin observed alterations in GR-associated gene expression. We also aimed to determine whether the methylated metabolites of iAs, namely monomethyl­arsenic (MMA) and dimethyl­arsenic (DMA), also influence GR-associated signaling in the placenta. The data indicate that iAs alters GR activation in a dose-dependent manner, reduces nuclear translocation, and reduces DNA binding. Additionally, the results demonstrate that MMA and DMA alter the expression of eight GR-associated genes, modulate GR activation, and alter DNA binding. These data are significant as they highlight the role of iAs as an endocrine disruptor and for the first time explore the effects of MMA and DMA on endocrine signaling in the placenta.


Assuntos
Arsênio/efeitos adversos , Arsenicais/efeitos adversos , Disruptores Endócrinos/efeitos adversos , Placenta/efeitos dos fármacos , Receptores de Glucocorticoides/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linhagem Celular Tumoral , Água Potável/efeitos adversos , Exposição Ambiental/efeitos adversos , Feminino , Humanos , Placenta/metabolismo , Gravidez
4.
Environ Toxicol ; 35(12): 1395-1405, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32790152

RESUMO

Per- and polyfluoroalkyl substances (PFAS), a class of environmental contaminants, have been detected in human placenta and cord blood. The mechanisms driving PFAS-induced effects on the placenta and adverse pregnancy outcomes are not well understood. This study investigated the impact of perfluorooctanesulfonic acid (PFOS), perfluorooctanoic acid (PFOA), and a replacement PFAS known as hexafluoropropylene oxide dimer acid (HFPO-DA, tradename GenX) on placental trophoblasts in vitro. Several key factors were addressed. First, PFAS levels in cell culture reagents at baseline were quantified. Second, the role of supplemental media serum in intracellular accumulation of PFAS in a human trophoblast (JEG3) cell line was established. Finally, the impact of PFAS on the expression of 96 genes involved in proper placental function in JEG3 cells was evaluated. The results revealed that serum-free media (SFM) contained no detectable PFAS. In contrast, fetal bovine serum-supplemented media (SSM) contained PFNA, PFUdA, PFTrDA, and 6:2 FTS, but these PFAS were not detected internally in cells. Intracellular accumulation following 24 hr treatments was significantly higher when cultured in SFM compared to SSM for PFOS and PFOA, but not HFPO-DA. Treatment with PFAS was associated with gene expression changes (n = 32) in pathways vital to placental function, including viability, syncytialization, inflammation, transport, and invasion/mesenchymal transition. Among the most robust PFAS-associated changes were those observed in the known apoptosis-related genes, BAD and BAX. These results suggest a complex relationship between PFAS, in vitro culture conditions, and altered expression of key genes necessary for proper placentation.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Caprilatos/toxicidade , Fluorocarbonos/toxicidade , Expressão Gênica/efeitos dos fármacos , Placenta/efeitos dos fármacos , Soro/química , Trofoblastos/efeitos dos fármacos , Ácidos Alcanossulfônicos/sangue , Ácidos Alcanossulfônicos/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/genética , Bioacumulação/efeitos dos fármacos , Bioacumulação/genética , Caprilatos/sangue , Caprilatos/metabolismo , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Meios de Cultura Livres de Soro , Feminino , Fluorocarbonos/sangue , Fluorocarbonos/metabolismo , Humanos , Placenta/metabolismo , Gravidez , RNA Mensageiro/genética , Trofoblastos/metabolismo
5.
Curr Environ Health Rep ; 7(3): 222-230, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32812200

RESUMO

PURPOSE OF REVIEW: This review summarizes studies highlighting perfluoroalkyl substances (PFAS) and their effects on the placenta, pregnancy outcomes, and child health. It highlights human population-based associations as well as in vitro-based experimental data to inform an understanding of the molecular mechanisms underlying these health effects. Among the mechanisms by which PFAS may induce toxicity is via their interaction with the peroxisome proliferator-activated receptors (PPARs), nuclear receptors that regulate lipid metabolism and placental functions important to healthy pregnancies, as well as fetal and child development. RECENT FINDINGS: In utero exposure to prevalent environmental contaminants such as PFAS is associated with negative health outcomes during pregnancy, birth outcomes, and later in life. Specifically, PFAS have been associated with increased incidence of gestational diabetes, childhood obesity, preeclampsia, and fetal growth restriction. In terms of placental molecular mechanisms underlying these associations, studies demonstrate that PFAS interfere with trophoblast lipid homeostasis, inflammation, and invasion. Moreover these effects could be mediated in part by the interaction between PFAS and PPARs, as well as other biological mechanisms. This review summarizes how PFAS, critical environmental contaminants, may contribute to diseases of pregnancy as well as early and later child health.


Assuntos
Desenvolvimento Infantil/efeitos dos fármacos , Exposição Ambiental/efeitos adversos , Fluorocarbonos/toxicidade , Exposição Materna/efeitos adversos , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Placenta/efeitos dos fármacos , Adulto , Criança , Saúde da Criança , Diabetes Gestacional/induzido quimicamente , Feminino , Feto/efeitos dos fármacos , Humanos , Gravidez , Resultado da Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente
6.
Artigo em Inglês | MEDLINE | ID: mdl-32210027

RESUMO

Air pollution consists of highly variable and complex mixtures recognized as major contributors to morbidity and mortality worldwide. The vast number of chemicals, coupled with limitations surrounding epidemiological and animal studies, has necessitated the development of new approach methods (NAMs) to evaluate air pollution toxicity. These alternative approaches include in vitro (cell-based) models, wherein toxicity of test atmospheres can be evaluated with increased efficiency compared to in vivo studies. In vitro exposure systems have recently been developed with the goal of evaluating air pollutant-induced toxicity; though the specific design parameters implemented in these NAMs-based studies remain in flux. This review aims to outline important design parameters to consider when using in vitro methods to evaluate air pollutant toxicity, with the goal of providing increased accuracy, reproducibility, and effectiveness when incorporating in vitro data into human health evaluations. This review is unique in that experimental considerations and lessons learned are provided, as gathered from first-hand experience developing and testing in vitro models coupled to exposure systems. Reviewed design aspects include cell models, cell exposure conditions, exposure chambers, and toxicity endpoints. Strategies are also discussed to incorporate in vitro findings into the context of in vivo toxicity and overall risk assessment.


Assuntos
Poluentes Atmosféricos/toxicidade , Poluição do Ar , Medição de Risco , Poluição do Ar/análise , Animais , Humanos , Reprodutibilidade dos Testes , Testes de Toxicidade
7.
Toxicol Sci ; 175(2): 210-219, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32219433

RESUMO

Per- and polyfluoroalkyl substances (PFAS) are used as industrial surfactants and chemical coatings for household goods such as Teflon. Despite regulatory efforts to phase out legacy PFAS, they remain detectable in drinking water throughout the United States. This is due to the stability of legacy PFAS and the continued use of replacement compounds. In humans, PFAS have been detected in placenta and cord blood and are associated with low birth weight and preeclampsia risk. Preeclampsia is a leading cause of maternal mortality and is driven by insufficient endometrial trophoblast invasion, resulting in poor placental blood flow. PFAS alter invasion of other cell types, but their impact on trophoblasts is not understood. We therefore assessed the effects of PFAS on trophoblast migration, invasion, and gene expression in vitro. Trophoblast migration and invasion were assessed using a modified scratch assay in the absence or presence of Matrigel, respectively. Treatment with perfluorooctanoic sulfate (PFOS), perfluorooctanoic acid (PFOA), and GenX (1000 ng/ml) each decreased trophoblast migration over 24 h. However, only GenX (1000 ng/ml) significantly inhibited trophoblast invasion. Treatment with PFOS, PFOA, and GenX also decreased trophoblast expression of chemokines (eg, CCL2), chemokine receptors (eg, CCR4), and inflammatory enzymes (eg, ALOX15) involved in migration. Inhibition of chemokine receptors with pertussis toxin (10 ng/ml), a G-protein inhibitor, inhibited trophoblast migration similar to the PFAS. Taken together, PFAS decrease trophoblast migration, invasion, and inflammatory signaling. By understanding the mechanisms involved, it may be possible to identify the biological and exposure factors that contribute to preeclampsia.


Assuntos
Circulação Sanguínea/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Água Potável/química , Fluorocarbonos/toxicidade , Pré-Eclâmpsia/induzido quimicamente , Tensoativos/toxicidade , Trofoblastos/efeitos dos fármacos , Adulto , Ensaios de Migração Celular , Feminino , Humanos , Gravidez , Estados Unidos
8.
Reprod Toxicol ; 98: 1-12, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32061676

RESUMO

Environmental chemicals comprise a major portion of the human exposome, with some shown to impact the health of susceptible populations, including pregnant women and developing fetuses. The placenta and cord blood serve as important biological windows into the maternal and fetal environments. In this article we review how environmental chemicals (defined here to include man-made chemicals [e.g., flame retardants, pesticides/herbicides, per- and polyfluoroalkyl substances], toxins, metals, and other xenobiotic compounds) contribute to the prenatal exposome and highlight future directions to advance this research field. Our findings from a survey of recent literature indicate the need to better understand the breadth of environmental chemicals that reach the placenta and cord blood, as well as the linkages between prenatal exposures, mechanisms of toxicity, and subsequent health outcomes. Research efforts tailored towards addressing these needs will provide a more comprehensive understanding of how environmental chemicals impact maternal and fetal health.


Assuntos
Expossoma , Desenvolvimento Fetal , Exposição Materna , Saúde Materna , Troca Materno-Fetal , Animais , Poluentes Ambientais/análise , Feminino , Sangue Fetal/química , Humanos , Placenta/química , Gravidez
9.
Toxicol Appl Pharmacol ; 378: 114635, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31233757

RESUMO

During pregnancy, the placenta is critical for the regulation of maternal homeostasis and fetal growth and development. Exposures to environmental chemicals during pregnancy can be detrimental to the health of the placenta and therefore adversely impact maternal and fetal health. Though research on placental-derived developmental toxicity is expanding, testing is limited by the resources required for traditional test methods based on whole animal experimentation. Alternative strategies utilizing in vitro methods are well suited to contribute to more efficient screening of chemical toxicity and identification of biological mechanisms underlying toxicity outcomes. This review aims to summarize methods that can be used to evaluate toxicity resulting from exposures during the prenatal period, with a focus on newer in vitro methods centered on placental toxicity. The following key aspects are reviewed: (i) traditional test methods based on animal developmental toxicity testing, (ii) in vitro methods using monocultures and explant models, as well as more recently developed methods, including co-cultures, placenta-on-a-chip, and 3-dimensional (3D) cell models, (iii) endpoints that are commonly measured using in vitro designs, and (iv) the translation of in vitro methods into chemical evaluations and risk assessment applications. We conclude that findings from in vitro placental models can contribute to the screening of potentially hazardous chemicals, elucidation of chemical mechanism of action, incorporation into adverse outcome pathways, estimation of doses eliciting toxicity, derivation of extrapolation factors, and characterization of overall risk of adverse outcomes, representing key components of chemical regulation in the 21st century.


Assuntos
Poluentes Ambientais/toxicidade , Desenvolvimento Fetal/efeitos dos fármacos , Substâncias Perigosas/toxicidade , Placenta/efeitos dos fármacos , Animais , Feminino , Humanos , Técnicas In Vitro/métodos , Gravidez , Medição de Risco , Testes de Toxicidade/métodos
10.
Chem Res Toxicol ; 32(3): 493-499, 2019 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-30746931

RESUMO

Prenatal exposure to inorganic arsenic (iAs) has been associated with adverse developmental and reproductive outcomes. These outcomes may be tied to altered functionality of nuclear transcription factors such as the glucocorticoid receptor (GR) in the placenta and associated gene expression. The GR pathway is integral for proper fetal and placental development, and perturbations in this pathway may underlie observed associations between prenatal iAs exposure and adverse birth outcomes. We therefore set out to investigate whether iAs modulates the GR signaling pathway in placental cells. JEG-3 trophoblasts were exposed to environmentally-relevant doses of iAs, and mRNA expression assessed. To examine the links between iAs exposure, the GR signaling pathway, and epigenetic modification, DNA methylation levels were also quantified. Treatment with iAs altered the expression of 12 GR-genes that play a role in fetal and placental development. Furthermore, at a gene-specific level, mRNA abundance was associated with changes in DNA methylation patterning in JEG-3 cells, suggesting that the effects of iAs are mediated by epigenetic mechanisms. The identified target genes have been associated with prenatal iAs exposure, placental physiology, and fetal development. This study provides further evidence for iAs as an endocrine disruptor and provides insight as to the mechanisms by which prenatal iAs exposure may induce adverse birth outcomes.


Assuntos
Arsênio/toxicidade , Ilhas de CpG/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Disruptores Endócrinos/toxicidade , Placenta/citologia , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Feminino , Humanos , Gravidez , Células Tumorais Cultivadas
11.
Pharmacol Res ; 141: 331-342, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30610963

RESUMO

The BCRP/ABCG2 efflux transporter is expressed on the membrane of placental syncytiotrophoblasts and protects the fetus from toxicant exposure. Syncytiotrophoblasts arise from the fusion of cytotrophoblasts, a process negatively regulated by the endocannabinoid, anandamide (AEA). It is unknown whether AEA can influence fetal concentrations of xenobiotics by modulating the expression of transporters in syncytiotrophoblasts. Here, we sought to characterize and identify the mechanism(s) responsible for AEA-mediated down-regulation of the BCRP transporter in human placental explants and BeWo trophoblasts. Treatment of human placental explants with AEA (1 µM, 24 h) reduced hCGα, syncytin-1, and BCRP mRNAs by ˜30%. Similarly, treatment of BeWo trophoblasts with AEA (0-10 µM, 3-24 h) coordinately down-regulated mRNAs for hCGß, syncytin-2, and BCRP. In turn, AEA increased the sensitivity of trophoblasts to the cytotoxicity of mitoxantrone, a known BCRP substrate, and environmental and dietary contaminants including mycoestrogens and perfluorinated chemicals. AEA-treated trophoblasts also demonstrated reduced BCRP transport of the mycoestrogen zearalenone and the diabetes drug glyburide, labeled with BODIPY. The AEA-mediated reduction of BCRP mRNA was abrogated when placental cells were co-treated with AM630, a CB2 receptor inhibitor, or 8-Br-cAMP, a cAMP analog. AEA reduced intracellular cAMP levels in trophoblasts by 75% at 1 h, and completely inhibited forskolin-induced phosphorylation of the cAMP response element binding protein (CREB). AEA also decreased p-CREB binding to the BCRP promoter. Taken together, our data indicate that AEA down-regulates placental transporter expression and activity via CB2-cAMP signaling. This novel mechanism may explain the repression of placental BCRP expression observed during diseases of pregnancy.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Ácidos Araquidônicos/farmacologia , Agonistas de Receptores de Canabinoides/farmacologia , AMP Cíclico/metabolismo , Regulação para Baixo/efeitos dos fármacos , Endocanabinoides/farmacologia , Proteínas de Neoplasias/genética , Placenta/efeitos dos fármacos , Alcamidas Poli-Insaturadas/farmacologia , Receptor CB2 de Canabinoide/metabolismo , Adulto , Linhagem Celular , Feminino , Humanos , Placenta/citologia , Placenta/metabolismo , Gravidez , Transdução de Sinais/efeitos dos fármacos , Trofoblastos/citologia , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo , Adulto Jovem
12.
Toxicol Sci ; 168(2): 394-404, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30576553

RESUMO

In the placenta, the breast cancer resistance protein (BCRP)/ABCG2 efflux transporter limits the maternal-to-fetal transfer of drugs and chemicals. Previous research has pointed to the estrogenic mycotoxin zearalenone as a potential substrate for BCRP. Here, we sought to assess the role of the BCRP transporter in the transplacental disposition of zearalenone during pregnancy. In vitro transwell transport assays employing BCRP/Bcrp-transfected Madine-Darby canine kidney cells and BeWo trophoblasts with reduced BCRP expression were used to characterize the impact of BCRP on the bidirectional transport of zearalenone. In both models, the presence of BCRP protein increased the basolateral-to-apical transport and reduced the apical-to-basolateral transport of zearalenone over a 2-h period. In vivo pharmacokinetic analyses were then performed using pregnant wild-type and Bcrp-/- mice after a single tail vein injection of zearalenone. Zearalenone and its metabolite α-zearalenol were detectable in serum, placentas, and fetuses from all animals, and ß-zearalenol was detected in serum and fetuses, but not placentas. There were no significant differences in the maternal serum concentrations of any analytes between the two genotypes. In Bcrp-/- mice, the free fetal concentrations of zearalenone, α-zearalenol, and ß-zearalenol were increased by 115%, 84%, and 150%, respectively, when compared with wild-type mice. Concentrations of free zearalenone and α-zearalenol were elevated 145% and 78% in Bcrp-/- placentas, respectively, when compared with wild-type placentas. Taken together, these data indicate that the placental BCRP transporter functions to reduce the fetal accumulation of zearalenone, which may impact susceptibility to developmental toxicities associated with in utero zearalenone exposure.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Estrogênios não Esteroides/farmacocinética , Feto/metabolismo , Troca Materno-Fetal/efeitos dos fármacos , Placenta/efeitos dos fármacos , Zearalenona/farmacocinética , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Animais , Transporte Biológico , Cães , Estrogênios não Esteroides/toxicidade , Feminino , Humanos , Células Madin Darby de Rim Canino , Troca Materno-Fetal/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Placenta/metabolismo , Gravidez , Distribuição Tecidual , Transfecção , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo , Zearalenona/toxicidade
13.
Toxicol Appl Pharmacol ; 359: 102-107, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30222979

RESUMO

NADH cytochrome b5 reductase mediates electron transfer from NADH to cytochrome b5 utilizing flavin adenine dinucleotide as a redox cofactor. Reduced cytochrome b5 is an important cofactor in many metabolic reactions including cytochrome P450-mediated xenobiotic metabolism, steroid biosynthesis and fatty acid metabolism, hemoglobin reduction, and methionine and plasmalogen synthesis. Using recombinant human enzyme, we discovered that cytochrome b5 reductase mediates redox cycling of a variety of quinones generating superoxide anion, hydrogen peroxide, and, in the presence of transition metals, hydroxyl radicals. Redox cycling activity was oxygen-dependent and preferentially utilized NADH as a co-substrate; NADH was 5-10 times more active than NADPH in supporting redox cycling. Redox cycling activity was greatest for 9,10-phenanthrenequinone and 1,2-naphthoquinone, followed by 1,4-naphthoquinone and 2-methyl-1,4-naphthoquinone (menadione), nitrofurantoin and 2-hydroxyestradiol. Using menadione as the substrate, quinone redox cycling was found to inhibit reduction of cytochrome b5 by cytochrome b5 reductase, as measured by heme spectral changes in cytochrome b5. Under anaerobic conditions where redox cycling is inhibited, menadione had no effect on the reduction of cytochrome b5. Chemical redox cycling by cytochrome b5 reductase may be important in generating cytotoxic reactive oxygen species in target tissues. This activity, together with the inhibition of cytochrome b5 reduction by redox-active chemicals and consequent deficiencies in available cellular cytochrome b5, are likely to contribute to tissue injury following exposure to quinones and related redox active chemicals.


Assuntos
Benzoquinonas/metabolismo , Citocromo-B(5) Redutase/metabolismo , Nitrofurantoína/metabolismo , Radicais Livres/metabolismo , Humanos , Cinética , Microssomos Hepáticos , NADP/metabolismo , Oxirredução , Consumo de Oxigênio , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/metabolismo
14.
Placenta ; 55: 29-36, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28623970

RESUMO

INTRODUCTION: The breast cancer resistance protein (BCRP/ABCG2) is an efflux transporter in the placental barrier. By transporting chemicals from the fetal to the maternal circulation, BCRP limits fetal exposure to a range of drugs, toxicants, and endobiotics such as bile acids and hormones. The purpose of the present studies was to 1) determine whether BCRP localizes to highly-ordered, cholesterol-rich lipid raft microdomains in placenta microvillous membranes, and 2) determine the impact of cholesterol on BCRP-mediated placental transport in vitro. METHODS: BCRP expression was analyzed in lipid rafts isolated from placentas from healthy, term pregnancies and BeWo trophoblasts by density gradient ultracentrifugation. BeWo cells were also tested for their ability to efflux BCRP substrates after treatment with the cholesterol sequestrant methyl-ß-cyclodextrin (MßCD, 5 mM, 1 h) or the cholesterol synthesis inhibitor pravastatin (200 µM, 48 h). RESULTS AND DISCUSSION: BCRP was found to co-localize with lipid raft proteins in detergent-resistant, lipid raft-containing fractions from placental microvillous membranes and BeWo cells. Treatment of BeWo cells with MßCD redistributed BCRP protein into higher density non-lipid raft fractions. Repletion of the cells with cholesterol restored BCRP localization to lipid raft-containing fractions. Treatment of BeWo cells with MßCD or pravastatin increased cellular retention of two BCRP substrates, the fluorescent dye Hoechst 33342 and the mycotoxin zearalenone. Repletion with cholesterol restored BCRP transporter activity. Taken together, these data demonstrate that cholesterol may play a critical role in the post-translational regulation of BCRP in placental lipid rafts.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Colesterol/metabolismo , Microdomínios da Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Placenta/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Gravidez , Processamento de Proteína Pós-Traducional
15.
Toxicol Sci ; 151(1): 150-9, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26880746

RESUMO

Cytochrome P450 (CYP) enzymes mediate mixed-function oxidation reactions important in drug metabolism. The aromatic heterocyclic cation, diphenyleneiodonium (DPI), binds flavin in cytochrome P450 reductase and inhibits CYP-mediated activity. DPI also inhibits CYP by directly interacting with heme. Herein, we report that DPI effectively inhibits a number of CYP-related monooxygenase reactions including NADPH oxidase, a microsomal enzyme activity that generates hydrogen peroxide in the absence of metabolizing substrates. Inhibition of monooxygenase by DPI was time and concentration dependent with IC50's ranging from 0.06 to 1.9 µM. Higher (4.6-23.9 µM), but not lower (0.06-1.9 µM), concentrations of DPI inhibited electron flow via cytochrome P450 reductase, as measured by its ability to reduce cytochrome c and mediate quinone redox cycling. Similar results were observed with inducible nitric oxide synthase (iNOS), an enzyme containing a C-terminal reductase domain homologous to cytochrome P450 reductase that mediates reduction of cytochrome c, and an N-terminal heme-thiolate oxygenase domain mediating nitric oxide production. Significantly greater concentrations of DPI were required to inhibit cytochrome c reduction by iNOS (IC50 = 3.5 µM) than nitric oxide production (IC50 = 0.16 µM). Difference spectra of liver microsomes, recombinant CYPs, and iNOS demonstrated that DPI altered heme-carbon monoxide interactions. In the presence of NADPH, DPI treatment of microsomes and iNOS yielded a type II spectral shift. These data indicate that DPI interacts with both flavin and heme in CYPs and iNOS. Increased sensitivity for inhibition of CYP-mediated metabolism and nitric oxide production by iNOS indicates that DPI targets heme moieties within the enzymes.


Assuntos
Inibidores das Enzimas do Citocromo P-450/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Heme/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Oniocompostos/farmacologia , Animais , Relação Dose-Resposta a Droga , Heme/metabolismo , Humanos , Camundongos , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos Sprague-Dawley , Proteínas Recombinantes/metabolismo , Fatores de Tempo
16.
Opt Express ; 20(24): 27147-54, 2012 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-23187570

RESUMO

A detailed study of the influence of the pulse duration, from the femtosecond to the nanosecond regime, on the evolution of the hole shape and depth during percussion drilling in silicon is presented. Real-time backlight imaging of the hole development is obtained for holes up to 2 mm deep with aspect ratios extending to 25:1. For low pulse energies, the hole-shape and drilling characteristics are similar for femtosecond, picoseconds and nanosecond regimes. At higher pulse energies, ns-pulses exhibit slower average drilling rates but eventually reach greater final depths. The shape of these holes is however dominated by branching and large internal cavities. For ps-pulses, a cylindrical shape is maintained with frequent small bulges on the side-walls. In contrast, fs-pulses cause only a limited number of imperfections on a tapered hole shape.


Assuntos
Interpretação de Imagem Assistida por Computador/métodos , Lasers , Fotometria/métodos , Teste de Materiais/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...