Network


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

Hotspot


Dive into the research topics where Gregory L. Kedderis is active.

Publication


Featured researches published by Gregory L. Kedderis.


Drug Metabolism Reviews | 1998

Sex-Dependent Metabolism of Xenobiotics

Cheryl A. Mugford; Gregory L. Kedderis

Sex-dependent differences in xenobiotic metabolism have been most extensively studied in the rat. Because sex-dependent differences are most pronounced in rats, this species quickly became the most popular animal model to study sexual dimorphisms in xenobiotic metabolism. Exaggerated sex-dependent variations in metabolism by rats may be the result of extensive inbreeding and/or differential evolution of isoforms of cytochromes P450 in mammals. For example, species-specific gene duplications and gene conversion events in the CYP2 and CYP3 families have produced different isoforms in rats and humans since the species division over 80 million years ago. This observation can help to explain the fact that CYP2C is not found in humans but is a major subfamily in rats (Table 11). Animal studies are used to help determine the metabolism and toxicity of many chemical agents in an attempt to extrapolate the risk of human exposure to these agents. One of the most important concepts in attempting to use rodent studies to identify sensitive individuals in the human population is that human cytochromes P450 differ from rodent cytochromes P450 in both isoform composition and catalytic activities. Xenobiotic metabolism by male rats can reflect human metabolism when the compound of interest is metabolized by CYP1A or CYP2E because there is strong regulatory conservation of these isoforms between rodents and humans. However, problems can arise when rats are used as animal models to predict the potential for sex-dependent differences in xenobiotic handling in humans. Information from countless studies has shown that the identification of sex-dependent differences in metabolism by rats does not translate across other animal species or humans. The major factor contributing to this observation is that CYP2C, a major subfamily in rats, which is expressed in a sex-specific manner, is not found in humans. To date, sex-specific isoforms of cytochromes P450 have not been identified in humans. The lack of expression of sex-dependent isoforms in humans indicates that the male rat is not an accurate model for the prediction of sex-dependent differences in humans. Differences in xenobiotic metabolism among humans are more likely the consequence of intraindividual variations as a result of genetics or environmental exposures rather than from sex-dependent differences in enzyme composition. A major component of the drug discovery and development process is to identify, at as early a stage as possible, the potential for toxicity in humans. Earlier identification of individual differences in xenobiotic metabolism and the potential for toxicity will be facilitated by improving techniques to make better use of human tissue to prepare accurate in vitro systems such as isolated hepatocytes and liver slices to study xenobiotic metabolism and drug-induced toxicities. Accurate systems should possess an array of bioactivation enzymes similar to the in vivo expression of human liver. In addition, the compound concentrations and exposure times used in these in vitro test systems should mimic those achieved in the target tissues of humans. Consideration of such factors will allow the development of compounds with improved efficacy and low toxicity at a more efficient rate. The development of accurate in vitro systems utilizing human tissue will also aid in the investigation of the molecular mechanisms by which the CYP genes are regulated in humans. Such studies will facilitate the study of the basis for differences in expression of isoforms of CYP450 in humans.


Chemico-Biological Interactions | 1999

Present status of the application of cryopreserved hepatocytes in the evaluation of xenobiotics: consensus of an international expert panel.

Albert P. Li; Peter D. Gorycki; Jan G. Hengstler; Gregory L. Kedderis; Hans G. Koebe; R. Rahmani; Georges de Sousas; José M. Silva; Paul Skett

Successful cryopreservation of freshly isolated hepatocytes would significantly decrease the need for freshly-procured livers for the preparation of hepatocytes for experimentation. Hepatocytes can be prepared, cryopreserved, and used for experimentation as needed at different times after isolation. Cryopreservation is especially important for research with human hepatocytes because of the limited availability of fresh human livers. Based on the cumulative experience of this international expert panel, a consensus was reached on the various aspects of hepatocyte cryopreservation, including cryopreservation and thawingprocedures and applications of the cryopreserved hepatocytes. Key to successful cryopreservation includes slow addition of cryopreservants, controlled-rate freezing with adjustment for the heat of crystallization, storage at -150 degrees C, and rapid thawing. There is a general consensus that cryopreserved hepatocytes are useful for short-term xenobiotic metabolism and cytotoxicity evaluation.


Chemico-Biological Interactions | 1997

Extrapolation of in vitro enzyme induction data to humans in vivo

Gregory L. Kedderis

Enzyme induction generally increases the rate and extent of xenobiotic metabolism in vitro, but physiological constraints can dampen these effects in vivo. Biotransformation kinetics determined in hepatocytes in vitro can be extrapolated to whole animals based on the hepatocellularity of the liver, since the initial velocity of an enzyme-catalyzed reaction is directly proportional to the total enzyme present in the cell. The biotransformation kinetics of various xenobiotics determined with isolated hepatocytes in vitro have been shown to accurately predict pharmacokinetics in whole animals. Analysis of the kinetic data, using physiologically based pharmacokinetics, allows extrapolation of xenobiotic biotransformation across dose routes and species in a biologically realistic context. Several fold variations were observed in the bioactivation of the hepatotoxicant furan by isolated human hepatocytes, due to induction of cytochrome P450 2E1. Extrapolation of these data to humans in vivo showed that furan bioactivation was limited by hepatic blood flow delivery of the substrate. One important consequence of hepatic blood flow limitation is that the amount of metabolite formed in the liver is unaffected by increases in Vmax due to enzyme induction. Therefore, interindividual variations in cytochrome P450 2E1 among human populations would not affect the bioactivation of many rapidly metabolized hazardous chemical air pollutants. The hepatic blood flow limitation of biotransformation is also observed after oral bolus dosing of rapidly metabolized compounds. More slowly metabolized xenobiotics, such as therapeutic agents, are only partially limited by hepatic blood flow and other processes.


Toxicology | 1997

Furan-induced liver cell proliferation and apoptosis in female B6C3F1 mice

Ronny Fransson-Steen; Thomas L. Goldsworthy; Gregory L. Kedderis; Robert R. Maronpot

Furan is a potent rodent hepatocarcinogen that probably acts through non-genotoxic mechanisms involving hepatotoxicity and regenerative hepatocyte proliferation. In addition to inducing necrosis, cytotoxicants like furan may also induce cytolethality through apoptosis which has been suggested to play a key role in carcinogenesis. Hepatocyte proliferation and apoptosis were studied in female B6C3F1 mice exposed to furan by oral gavage for 3 weeks at National Toxicology Program (NTP) bioassay doses (8 and 15 mg/kg body weight) and lower (4 mg/kg). Furan treatment led to a 2- to 3-fold significant increase in liver-related enzymes and bile acids in blood serum as compared to the control group. These changes were accompanied by minor subcapsular inflammation and minimal necrosis at 8 and 15 mg furan/kg. A dose-related increase in bromodeoxyuridine-labeling index (1.4- to 1.7-fold) and hematoxylin- and eosin-defined apoptotic index (6- to 15-fold) was observed at 8 and 15 mg/kg. Co-treatment of mice with aminobenzotriazole, an irreversible inhibitor of cytochromes P-450, prevented the observed hepatotoxic effects induced by furan. These results indicate that furan elicits hepatotoxicity in a dose-related manner through a toxic metabolite and, furthermore, suggest that apoptosis is an important form of cell death at hepatocarinogenic doses under short-term conditions.


Science of The Total Environment | 2002

Incorporating human interindividual biotransformation variance in health risk assessment.

John C. Lipscomb; Gregory L. Kedderis

The protection of sensitive individuals within a population dictates that measures other than central tendencies be employed to estimate risk. The refinement of human health risk assessments for chemicals metabolized by the liver to reflect data on human variability can be accomplished through (1) the characterization of enzyme expression in large banks of human liver samples, (2) the employment of appropriate techniques for the quantification and extrapolation of metabolic rates derived in vitro, and (3) the judicious application of physiologically based pharmacokinetic (PBPK) modeling. While in vitro measurements of specific biochemical reactions from multiple human samples can yield qualitatively valuable data on human variance, such measures must be put into the perspective of the intact human to yield the most valuable predictions of metabolic differences among humans. For quantitative metabolism data to be the most valuable in risk assessment, they must be tied to human anatomy and physiology, and the impact of their variance evaluated under real exposure scenarios. For chemicals metabolized in the liver, the concentration of parent chemical in the liver represents the substrate concentration in the Michaelis Menten description of metabolism. Metabolic constants derived in vitro may be extrapolated to the intact liver, when appropriate conditions are met. Metabolic capacity Vmax; the maximal rate of the reaction) can be scaled directly to the concentration of enzyme (or enzyme fraction) contained in the liver. Several environmental, genetic and lifestyle factors can influence the concentration of cytochrome P450 forms (CYP) in the liver by affecting either (1) the extent to which the CYP forms are expressed in the endoplasmic reticulum of the cell (isolated as the microsomal fraction from tissue homogenates), or (2) the expression of microsomal protein in intact liver tissue. Biochemically sound measures of the hepatic distribution of xenobiotic metabolizing enzymes among humans, based on expression of the enzymes within microsomal protein and the distribution of microsomal protein among intact livers, can be combined with metabolic constants derived in vitro to generate values consistent with those employed in PBPK models. When completed, the distribution (and bounds) of Vmax values can be estimated and included in PBPK models. Exercising such models under plausible exposure scenarios will demonstrate the extent to which human interindividual enzyme variance can influence parameters (i.e., the detoxication of a toxic chemical through metabolism) that may influence risk. In this article, we describe a methodology and conditions which must exist for such an approach to be successful.


Toxicology Letters | 1995

The role of regenerative cell proliferation in chloroform- induced cancer

Byron E. Butterworth; Michael V. Templin; Susan J. Borghoff; Rory B. Conolly; Gregory L. Kedderis; Douglas C. Wolf

Chloroform produces cancer by a nongenotoxic-cytotoxic mode of action, with no increased cancer risk expected at noncytotoxic doses. The default risk assessment for inhaled chloroform relies on liver tumor incidence from a gavage study with female B6C3F1 mice and estimates a virtually safe dose (VSD) at an airborne concentration of 0.000008 ppm of chloroform. In contrast, a 1000-fold safety factor applied to the NOAEL for liver cytotoxicity from inhalation studies yields a VSD of 0.01 ppm. This estimate relies on inhalation data and is more consistent with the mode of action of chloroform.


Risk Analysis | 2000

Improving Cancer Dose–Response Characterization by Using Physiologically Based Pharmacokinetic Modeling: An Analysis of Pooled Data for Acrylonitrile‐Induced Brain Tumors to Assess Cancer Potency in the Rat

Christopher R. Kirman; Sean M. Hays; Gregory L. Kedderis; Michael L. Gargas; Dale E. Strother

Historically, U.S. regulators have derived cancer slope factors by using applied dose and tumor response data from a single key bioassay or by averaging the cancer slope factors of several key bioassays. Recent changes in U.S. Environmental Protection Agency (EPA) guidelines for cancer risk assessment have acknowledged the value of better use of mechanistic data and better dose-response characterization. However, agency guidelines may benefit from additional considerations presented in this paper. An exploratory study was conducted by using rat brain tumor data for acrylonitrile (AN) to investigate the use of physiologically based pharmacokinetic (PBPK) modeling along with pooling of dose-response data across routes of exposure as a means for improving carcinogen risk assessment methods. In this study, two contrasting assessments were conducted for AN-induced brain tumors in the rat on the basis of (1) the EPAs approach, the dose-response relationship was characterized by using administered dose/concentration for each of the key studies assessed individually; and (2) an analysis of the pooled data, the dose-response relationship was characterized by using PBPK-derived internal dose measures for a combined database of ten bioassays. The cancer potencies predicted for AN by the contrasting assessments are remarkably different (i.e., risk-specific doses differ by as much as two to four orders of magnitude), with the pooled data assessments yielding lower values. This result suggests that current carcinogen risk assessment practices overestimate AN cancer potency. This methodology should be equally applicable to other data-rich chemicals in identifying (1) a useful dose measure, (2) an appropriate dose-response model, (3) an acceptable point of departure, and (4) an appropriate method of extrapolation from the range of observation to the range of prediction when a chemicals mode of action remains uncertain.


Biochemical Pharmacology | 1991

Glutathione S-transferase-mediated chlorothalonil metabolism in liver and gill subcellular fractions of channel catfish.

Evan P. Gallagher; Gregory L. Kedderis; Richard T. Di Giulio

Chlorothalonil (2,4,5,6-tetrachloroisophthalonitrile) is a broad spectrum fungicide that is a potent acute toxicant to fish. Therefore, the metabolism of chlorothalonil was investigated in liver and gill cytosolic and microsomal fractions from channel catfish (Ictalurus punctatus) using HPLC. All fractions catalyzed the metabolism of chlorothalonil to polar metabolites. Chlorothalonil metabolism by cytosolic fractions was reduced markedly when glutathione (GSH) was omitted from the reaction mixtures. The lack of microsomal metabolism in the presence of either NADPH or an NADPH-regenerating system indicated direct glutathione S-transferase (GST)-catalyzed conjugation with GSH without prior oxidation by cytochrome P450. Cytosolic and microsomal GSTs from both tissues were also active toward 1-chloro-2,4-dinitrobenzene (CDNB), a commonly employed reference substrate. In summary, channel catfish detoxified chlorothalonil in vitro by GST-catalyzed GSH conjugation in the liver and gill. The present report is the first to confirm microsomal GST activity toward CDNB in gill and toward chlorothalonil in liver, and also of gill cytosolic GST activity towards chlorothalonil, in an aquatic species.


Toxicology and Industrial Health | 2001

Application of in vitro biotransformation data and pharmacokinetic modeling to risk assessment

Gregory L. Kedderis; John C. Lipscomb

The adverse biological effects of toxic substances are dependent upon the exposure concentration and the duration of exposure. Pharmacokinetic models can quantitatively relate the external concentration of a toxicant in the environment to the internal dose of the toxicant in the target tissues of an exposed organism. The exposure concentration of a toxic substance is usually not the same as the concentration of the active form of the toxicant that reaches the target tissues following absorption, distribution, and biotransformation of the parent toxicant. Biotransformation modulates the biological activity of chemicals through bioactivation and detoxication pathways. Many toxicants require biotransformation to exert their adverse biological effects. Considerable species differences in biotransformation and other pharmacokinetic processes can make extrapolation of toxicity data from laboratory animals to humans problematic. Additionally, interindividual differences in biotransformation among human populations with diverse genetics and lifestyles can lead to considerable variability in the bioactivation of toxic chemicals. Compartmental pharmacokinetic models of animals and humans are needed to understand the quantitative relationships between chemical exposure and target tissue dose as well as animal to human differences and interindividual differences in human populations. The data-based compartmental pharmacokinetic models widely used in clinical pharmacology have little utility for human health risk assessment because they cannot extrapolate across dose route or species. Physiologically based pharmacokinetic (PBPK) models allow such extrapolations because they are based on anatomy, physiology, and biochemistry. In PBPK models, the compartments represent organs or groups of organs and the flows between compartments are actual blood flows. The concentration of a toxicant in a target tissue is a function of the solubility of the toxicant in blood and tissues (partition coefficients), blood flow into the tissue, metabolism of the toxicant in the tissue, and blood flow out of the tissue. The appropriate degree of biochemical detail can be added to the PBPK models as needed. Comparison of model simulations with experimental data provides a means of hypothesis testing and model refinement. In vitro biotransformation data from studies with isolated liver cells or subcellular fractions from animals or humans can be extrapolated to the intact organism based upon protein content or cell number. In vitro biotransformation studies with human liver preparations can provide quantitative data on human interindividual differences in chemical bioactivation. These in vitro data must be integrated into physiological models to understand the true impact of interindividual differences in chemical biotransformation on the target organ bioactivation of chemical contaminants in air and drinking water.


Chemico-Biological Interactions | 1984

Hepatic macromolecular covalent binding of mononitrotoluenes in Fischer-344 rats

Douglas E. Rickert; Rochelle M. Long; Martin C. Dyroff; Gregory L. Kedderis

The mononitrotoluenes are important industrial chemicals which display isomeric specificity in their ability to induce hepatic DNA excision repair in Fischer-344 rats. Covalent binding of the structurally related hepatocarcinogen, 2,6-dinitrotoluene, to hepatic DNA is markedly decreased by prior administration of the sulfotransferase inhibitors pentachlorophenol (PCP) and 2,6-dichloro-4-nitrophenol (DCNP). The objectives of this study were to determine whether hepatic macromolecular covalent binding of the mononitrotoluene isomers differed and to determine whether covalent binding of the mononitrotoluenes to hepatic DNA in vivo was decreased by inhibitors of sulfotransferase. Male Fischer-344 rats were given a single oral dose of [ring-U-14C]-2-, 3- or 4-nitrotoluene (2-, 3- or 4-NT) and killed at various times thereafter. Livers were removed and analyzed for total and covalently bound radiolabel. Maximal concentrations of total radiolabel were observed between 3 and 12 h after the dose, and there were no large differences among the 3 isomers in peak concentrations achieved. Covalent binding to hepatic macromolecules was maximal 12 h after administration for all three isomers. Thereafter, concentrations of covalently bound 2-NT-derived material were always 2-6 times higher than those of 3- or 4-NT-derived material. When DNA was isolated from livers of rats given the mononitrotoluenes 12 h previously, only 2-NT was observed to covalently bind at concentrations above the limits of detection of the assay. The covalent binding of 2-NT, but not that of 3- or 4-NT, to both total hepatic macromolecules and DNA was markedly decreased by prior administration of either PCP or DCNP. Covalent binding to hepatic DNA was decreased by greater than 96%. The results of this study correlate well with studies which have demonstrated that 2-NT, but not 3- or 4-NT, induces DNA excision repair. Furthermore, they suggest that 2-NT, like the hepatocarcinogen 2,6-dinitrotoluene, requires the action of sulfotransferase for its conversion to a species capable of covalently binding to hepatic DNA.

Collaboration


Dive into the Gregory L. Kedderis's collaboration.

Top Co-Authors

Avatar

Renu Batra

Research Triangle Park

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John C. Lipscomb

United States Environmental Protection Agency

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge