Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Gabriel A. Knudsen is active.

Publication


Featured researches published by Gabriel A. Knudsen.


Environmental Health Perspectives | 2013

Mimicking of Estradiol Binding by Flame Retardants and Their Metabolites: A Crystallographic Analysis.

Rajendrakumar A. Gosavi; Gabriel A. Knudsen; Linda S. Birnbaum; Lars C. Pedersen

Background: Brominated flame retardants (BFRs), used in many types of consumer goods, are being studied because of concerns about possible health effects related to endocrine disruption, immunotoxicity, reproductive toxicity, and neurotoxicity. Tetrabromobisphenol A (TBBPA), the most widely used BFR, and human metabolites of certain congeners of polybrominated diphenyl ether (e.g., 3-OH-BDE-47) have been suggested to inhibit estrogen sulfotransferase, potentially affecting estrogen metabolism. Objectives: Our primary goal was to understand the structural mechanism for inhibition of the hormone-metabolizing enzyme estrogen sulfotransferase by certain BFRs. We also sought to understand various factors that facilitate the binding of flame retardants in the enzyme binding pocket. Methods: We used X-ray crystallography to obtain atomic detail of the binding modes of TBBPA and 3-OH-BDE-47 to estrogen sulfotransferase for comparison with binding of the endogenous substrate estradiol. Results: The crystal structures reveal how BFRs mimic estradiol binding as well as the various interactions between the compounds and protein residues that facilitate its binding. In addition, the structures provide insights into the ability of the sulfotransferase substrate binding pocket to accommodate a range of halogenated compounds that satisfy minimal structural criteria. Conclusions: Our results show how BFRs or their metabolites can bind to and inhibit a key hormone-metabolizing enzyme, potentially causing endocrine disruption. Citation: Gosavi RA, Knudsen GA, Birnbaum LS, Pedersen LC. 2013. Mimicking of estradiol binding by flame retardants and their metabolites: a crystallographic analysis. Environ Health Perspect 121:1194–1199; http://dx.doi.org/10.1289/ehp.1306902


Environmental Health Perspectives | 2014

Diversity Outbred Mice Identify Population-Based Exposure Thresholds and Genetic Factors that Influence Benzene-Induced Genotoxicity

John E. French; Daniel M. Gatti; Daniel L. Morgan; Grace E. Kissling; Keith R. Shockley; Gabriel A. Knudsen; Kim G. Shepard; Herman C. Price; Deborah King; Kristine L. Witt; Lars C. Pedersen; Steven C. Munger; Karen L. Svenson; Gary A. Churchill

Background Inhalation of benzene at levels below the current exposure limit values leads to hematotoxicity in occupationally exposed workers. Objective We sought to evaluate Diversity Outbred (DO) mice as a tool for exposure threshold assessment and to identify genetic factors that influence benzene-induced genotoxicity. Methods We exposed male DO mice to benzene (0, 1, 10, or 100 ppm; 75 mice/exposure group) via inhalation for 28 days (6 hr/day for 5 days/week). The study was repeated using two independent cohorts of 300 animals each. We measured micronuclei frequency in reticulocytes from peripheral blood and bone marrow and applied benchmark concentration modeling to estimate exposure thresholds. We genotyped the mice and performed linkage analysis. Results We observed a dose-dependent increase in benzene-induced chromosomal damage and estimated a benchmark concentration limit of 0.205 ppm benzene using DO mice. This estimate is an order of magnitude below the value estimated using B6C3F1 mice. We identified a locus on Chr 10 (31.87 Mb) that contained a pair of overexpressed sulfotransferases that were inversely correlated with genotoxicity. Conclusions The genetically diverse DO mice provided a reproducible response to benzene exposure. The DO mice display interindividual variation in toxicity response and, as such, may more accurately reflect the range of response that is observed in human populations. Studies using DO mice can localize genetic associations with high precision. The identification of sulfotransferases as candidate genes suggests that DO mice may provide additional insight into benzene-induced genotoxicity. Citation French JE, Gatti DM, Morgan DL, Kissling GE, Shockley KR, Knudsen GA, Shepard KG, Price HC, King D, Witt KL, Pedersen LC, Munger SC, Svenson KL, Churchill GA. 2015. Diversity Outbred mice identify population-based exposure thresholds and genetic factors that influence benzene-induced genotoxicity. Environ Health Perspect 123:237–245; http://dx.doi.org/10.1289/ehp.1408202


Toxicology reports | 2014

Disposition and kinetics of tetrabromobisphenol A in female Wistar Han rats

Gabriel A. Knudsen; J. Michael Sanders; Abdella M. Sadik; Linda S. Birnbaum

Tetrabromobisphenol A (TBBPA) is the brominated flame retardant with the largest production volume worldwide. NTP 2-year bioassays found TBBPA dose-dependent increases in uterine tumors in female Wistar Han rats; evidence of reproductive tissues carcinogenicity was equivocal in male rats. To explain this apparent sex-dependence, the disposition and toxicokinetic profile of TBBPA were investigated using female Wistar Han rats, as no data were available for female rats. In these studies, the primary route of elimination following [14C]-TBBPA administration (25, 250 or 1000 mg/kg) was in feces; recoveries in 72 h were 95.7 ± 3.5%, 94.3 ± 3.6% and 98.8 ± 2.2%, respectively (urine: 0.2–2%; tissues: <0.1). TBBPA was conjugated to mono-glucuronide and -sulfate metabolites and eliminated in the bile. Plasma toxicokinetic parameters for a 250 mg/kg dose were estimated based on free TBBPA, as determined by UV/radiometric-HPLC analyses. Oral dosing by gavage (250 mg/kg) resulted in a rapid absorption of compound into the systemic circulation with an observed Cmax at 1.5 h post-dose followed by a prolonged terminal phase. TBBPA concentrations in plasma decreased rapidly after an IV dose (25 mg/kg) followed by a long elimination phase. These results indicate low systemic bioavailability (F < 0.05), similar to previous reports using male rats. Elimination pathways appeared to become saturated leading to delayed excretion after a single oral administration of the highest dose (1000 mg/kg); no such saturation or delay was detected at lower doses. Chronic high exposures to TBBPA may result in competition for metabolism with endogenous substrates in extrahepatic tissues (e.g., SULT1E1 estrogen sulfation) resulting in endocrine disruption.


Toxicology and Applied Pharmacology | 2016

Disruption of estrogen homeostasis as a mechanism for uterine toxicity in Wistar Han rats treated with tetrabromobisphenol A

J. Michael Sanders; Sherry J. Coulter; Gabriel A. Knudsen; June K. Dunnick; Grace E. Kissling; Linda S. Birnbaum

Chronic oral treatment of tetrabromobisphenol A (TBBPA) to female Wistar Han rats resulted in increased incidence of cell proliferation at 250mg/kg and tumor formation in the uterus at higher doses. The present study was designed to test the hypothesis that disruption of estrogen homeostasis was a major mode-of-action for the observed effects. Biological changes were assessed in serum, liver, and the proximal (nearest the cervix) and distal (nearest the ovaries) sections of the uterine horn of Wistar Han rats 24h following administration of the last of five daily oral doses of 250mg/kg. Expression of genes associated with receptors, biosynthesis, and metabolism of estrogen was altered in the liver and uterus. TBBPA treatment also resulted in changes in expression of genes associated with cell division and growth. Changes were also observed in the concentration of thyroxine in serum and in expression of genes in the liver and uterus associated with thyroid hormone receptors. Differential expression of some genes was tissue-dependent or specific to tissue location in the uterus. The biological responses observed in the present study support the hypothesis that perturbation of estrogen homeostasis is a major mode-of-action for TBBPA-mediated cell proliferation and tumorigenesis previously observed in the uterus of TBBPA-treated Wistar Han rats.


Toxicology and Applied Pharmacology | 2015

Estimation of tetrabromobisphenol A (TBBPA) percutaneous uptake in humans using the parallelogram method.

Gabriel A. Knudsen; Michael F. Hughes; Katelyn L. McIntosh; J. Michael Sanders; Linda S. Birnbaum

Tetrabromobisphenol A (TBBPA) is currently the worlds highest production volume brominated flame retardant. Humans are frequently exposed to TBBPA by the dermal route. In the present study, a parallelogram approach was used to make predictions of internal dose in exposed humans. Human and rat skin samples received 100 nmol of TBBPA/cm(2) skin and absorption and penetrance were determined using a flow-through in vitro system. TBBPA-derived [(14)C]-radioactivity was determined at 6h intervals in the media and at 24h post-dosing in the skin. The human skin and media contained an average of 3.4% and 0.2% of the total dose at the terminal time point, respectively, while the rat skin and media contained 9.3% and 3.5%, respectively. In the intact rat, 14% of a dermally-administered dose of ~100 nmol/cm(2) remained in the skin at the dosing site, with an additional 8% reaching systemic circulation by 24h post-dosing. Relative absorption and penetrance were less (10% total) at 24h following dermal administration of a ten-fold higher dose (~1000 nmol/cm(2)) to rats. However, by 72 h, 70% of this dose was either absorbed into the dosing-site skin or had reached systemic circulation. It is clear from these results that TBBPA can be absorbed by the skin and dermal contact with TBBPA may represent a small but important route of exposure. Together, these in vitro data in human and rat skin and in vivo data from rats may be used to predict TBBPA absorption in humans following dermal exposure. Based on this parallelogram calculation, up to 6% of dermally applied TBBPA may be bioavailable to humans exposed to TBBPA.


Toxicological Sciences | 2013

The Fate of β-Hexabromocyclododecane in Female C57BL/6 Mice

J. Michael Sanders; Gabriel A. Knudsen; Linda S. Birnbaum

1,2,5,6,9,10-Hexabromocyclododecane (HBCD) is a high production volume cycloaliphatic used as an additive flame retardant primarily in polystyrene foam building materials. HBCD mixtures contain three major stereoisomers, alpha (α), beta (β), and gamma (γ), at a typical ratio of 1.2:0.6:8.2. The toxicokinetic properties of the α and γ isomers differ. For instance, α-HBCD has greater bioavailability and potential for accumulation in mice than γ-HBCD. The present study reports comparative kinetics data for β-HBCD needed to support toxicological evaluations of HBCD mixtures. Results indicated that a single oral dose of 3mg/kg of [(14)C]-labeled β-HBCD was absorbed rapidly (≥ 85% total dose) in the female C57BL/6 mouse. The C max for β-HBCD-derived radioactivity in tissues, except adipose, was observed 3h following gavage. Approximately 90% of the administered dose was excreted in urine and feces within 24h, primarily as β-HBCD-derived metabolites. A portion of the dose (circa 9%) was excreted in feces as γ-HBCD. Oral administration of 30 or 100mg/kg of β-HBCD resulted initially in slower rates of [(14)C] elimination; however, cumulative excretion data were similar across the dosing range 4 days postdosing. Residual concentrations of [(14)C] in tissues were highest in adipose and liver. β-HBCD-derived radioactivity accumulated in most tissues following four consecutive daily oral doses of 3mg/kg. The extent of metabolism and excretion of β-HBCD in female C57BL/6 mice was similar to that for γ-HBCD. The potential for accumulation of β-HBCD-derived material in most tissues appeared to be less than for α-HBCD.


Xenobiotica | 2017

Disposition of the emerging brominated flame retardant, bis(2-ethylhexyl) tetrabromophthalate, in female Sprague Dawley rats: effects of dose, route and repeated administration

Gabriel A. Knudsen; J. Michael Sanders; Linda S. Birnbaum

Abstract 1. Bis(2-ethylhexyl)-tetrabromophthalate (BEH-TEBP; CAS No. 26040-51-7; PubChem CID: 117291; MW 706.15 g/mol, elsewhere: TeBrDEPH, TBPH, or BEHTBP) is used as an additive brominated flame retardant in consumer products. 2. Female Sprague Dawley rats eliminated 92–98% of [14C]-BEH-TEBP unchanged in feces after oral administration (0.1 or 10 μmol/kg). A minor amount of each dose (0.8–1%) was found in urine after 72 h. Disposition of orally administered BEH-TEBP in male B6C3F1/Tac mice was similar to female rats. 3. Bioaccumulation of [14C]-radioactivity was observed in liver and adrenals following 10 daily oral administrations (0.1 μmol/kg/day). These tissues contained 5- and 10-fold higher concentrations of [14C]-radioactivity, respectively, versus a single dose. 4. IV-administered [14C]-BEH-TEBP (0.1 μmol/kg) was slowly eliminated in feces, with >15% retained in tissues after 72 h. Bile and fecal extracts from these rats contained the metabolite mono-ethylhexyl tetrabromophthalate (TBMEHP). 5. BEH-TEBP was poorly absorbed, minimally metabolized and eliminated mostly by the fecal route after oral administration. Repeated exposure to BEH-TEBP led to accumulation in some tissues. The toxicological significance of this effect remains to be determined. This work was supported by the Intramural Research Program of the National Cancer Institute at the National Institutes of Health (Project ZIA BC 011476).


Toxicology Letters | 2017

Gene expression changes in immune response pathways following oral administration of tetrabromobisphenol A (TBBPA) in female Wistar Han rats

Samantha M. Hall; Sherry J. Coulter; Gabriel A. Knudsen; J. Michael Sanders; Linda S. Birnbaum

Tetrabromobisphenol A (TBBPA) is a brominated flame retardant used globally at high volumes, primarily in the epoxy resin of circuit boards. It has been detected in the environment and in humans. The National Toxicology Program found that chronic oral TBBPA treatment of 250mg/kg and higher caused an increased incidence of uterine lesions in female Wistar Han rats. The present laboratory has previously reported changes in gene expression associated with estrogen homeostasis in liver and uterine tissue of adult female Wistar Han rats after five days of gavage with 250mg/kg of TBBPA. Microarray analysis of tissue from these same TBBPA-treated rats was performed to detect additional pathways perturbed by TBBPA. Microarray analysis of uterine tissue detected downregulation of genes in pathways of the immune response following TBBPA treatment. These results, along with validation of associated gene expression changes using droplet digital PCR, are reported here. Our findings suggest mechanisms that may be related to estrogen-mediated immunosuppression.


Xenobiotica | 2017

The biological fate of decabromodiphenyl ethane following oral, dermal or intravenous administration

Gabriel A. Knudsen; J. Michael Sanders; Michael F. Hughes; Ethan P. Hull; Linda S. Birnbaum

Abstract 1. It was important to investigate the disposition of decabromodiphenyl ethane (DBDPE) based on concerns over its structural similarities to decabromodiphenyl ether (decaBDE), high potential for environmental persistence and bioaccumulation, and high production volume. 2. In the present study, female Sprague Dawley rats were administered a single dose of [14C]-DBDPE by oral, topical or IV routes. Another set of rats were administered 10 daily oral doses of [14C]-DBDPE. Male B6C3F1/Tac mice were administered a single oral dose. 3. DBDPE was poorly absorbed following oral dosing, with 95% of administered [14C]-radioactivity recovered in the feces unchanged, 1% recovered in the urine and less than 3% in the tissues at 72 h. DBDPE excretion was similar in male mice and female rats. Accumulation of [14C]-DBDPE was observed in liver and the adrenal gland after 10 daily oral doses to rats. 4. Rat and human skin were used to assess potential dermal uptake of DBDPE. The dermis was a depot for dermally applied DBDPE; conservative estimates predict ∼14 ± 8% of DBDPE may be absorbed into human skin in vivo; ∼7 ± 4% of the parent chemical is expected to reach systemic circulation following continuous exposure (24 h). 5. Following intravenous administration, ∼70% of the dose remained in tissues after 72 h, with the highest concentrations found in lung (1223 ± 723 pmol-eq/g), spleen (1096 ± 369 pmol-eq/g) and liver (366 ± 98 pmol-eq/g); 5 ± 1% of the dose was recovered in urine and 26 ± 4% in the feces.


Environmental Health Perspectives | 2014

Crystallographic analysis and mimicking of estradiol binding: Pedersen et al. Respond.

Lars C. Pedersen; Linda S. Birnbaum; Rajendrakumar A. Gosavi; Gabriel A. Knudsen

Previous studies have addressed the biological effects of brominated flame retardants (Birnbaum and Staskal 2004; Koike et al. 2013; Mariussen and Fonnum 2003; Ogunbayo et al. 2008), including a 2-year bioassay study performed by the National Toxicology Program (NTP), which demonstrated that tetrabromobisphenol A (TBBPA) can induce aggressive uterine tumors in rats (NTP 2013). As pointed out by Osimitz et al., TBBPA has been shown to bind poorly to the estrogen receptor, providing the impetus to study other pathways such as disruption of steroid transport and metabolism. Other groups have demonstrated the ability of TBBPA and flame retardant metabolites to inhibit estrogen sulfotransferase (SULT1E1), with IC50 (median inhibitory concentration) values near the Km for estradiol (Hamers et al. 2008; Kester et al. 2002; Zhang et al. 1998). Our work (Gosavi et al. 2013) was focused solely on understanding the structural mechanism by which these compounds bind to and inhibit SULT1E1’s ability to metabolize estradiol. The results of our work demonstrate that TBBPA and the 3-OH metabolite of BDE-47, although structurally different, bind in a similar manner at the estradiol binding site. This work suggests that these compounds could have an additive effect on the inhibition of this enzyme. We wholeheartedly agree with Osimitz et al. that the results of our work warrant future studies addressing the potential additive effect of these compounds on steroid metabolism in target tissues.

Collaboration


Dive into the Gabriel A. Knudsen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lars C. Pedersen

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Grace E. Kissling

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Michael F. Hughes

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel L. Morgan

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Daniel M. Gatti

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Herman C. Price

Alion Science and Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge