Network


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

Hotspot


Dive into the research topics where Randy L. Rose is active.

Publication


Featured researches published by Randy L. Rose.


Chemico-Biological Interactions | 2002

In vitro metabolism of carbaryl by human cytochrome P450 and its inhibition by chlorpyrifos.

Jun Tang; Yan Cao; Randy L. Rose; Ernest Hodgson

Carbaryl is a widely used anticholinesterase carbamate insecticide. Although previous studies have demonstrated that carbaryl can be metabolized by cytochrome P450 (CYP), the identification and characterization of CYP isoforms involved in metabolism have not been described either in humans or in experimental animals. The in vitro metabolic activities of human liver microsomes (HLM) and human cytochrome P450 (CYP) isoforms toward carbaryl were investigated in this study. The three major metabolites, i.e. 5-hydroxycarbaryl, 4-hydroxycarbaryl and carbaryl methylol, were identified after incubation of carbaryl with HLM or individual CYP isoforms and analysis by HPLC. Most of the 16 human CYP isoforms studied showed some metabolic activity toward carbaryl. CYP1A1 and 1A2 had the greatest ability to form 5-hydroxycarbaryl, while CYP3A4 and CYP1A1 were the most active in generation of 4-hydroxycarbaryl. The production of carbaryl methylol was primarily the result of metabolism by CYP2B6. Differential activities toward carbaryl were observed among five selected individual HLM samples with the largest difference occurring in the production of carbaryl methylol. Co-incubations of carbaryl and chlorpyrifos in HLM greatly inhibited carbaryl metabolism. The ability of HLM to metabolize carbaryl was also reduced by pre-incubation of HLM with chlorpyrifos. Chlorpyrifos inhibited the generation of carbaryl methylol, catalyzed predominately by CYP2B6, more than other pathways, correlating with an earlier observation that chlorpyrifos is metabolized to its oxon primarily by CYP2B6. Therefore, carbaryl metabolism in humans and its interaction with other chemicals is reflected by the concentration of CYP isoforms in HLM and their activities in the metabolic pathways for carbaryl. (Supported by NCDA Environmental Trust Fund)


Environmental Health Perspectives | 2000

Comparative metabolism of chloroacetamide herbicides and selected metabolites in human and rat liver microsomes.

Scott Coleman; Russell J. Linderman; Ernest Hodgson; Randy L. Rose

Acetochlor [2-chloro-N-(ethoxymethyl)-N-(2-ethyl-6-methyl-phenyl)-acetamide], alachlor [N-(methoxymethyl)-2-chloro-N-(2, 6-diethyl-phenyl)acetamide], butachlor [N-(butoxymethyl)-2-chloro-N-(2,6-diethyl-phenyl)acetamide], and metolachlor [2-chloro-N-(2-ethyl-6-methylphenyl)-N-(2-methoxy-1-methylethyl) acetamide] are pre-emergent herbicides used in the production of agricultural crops. These herbicides are carcinogenic in rats: acetochlor and alachlor cause tumors in the nasal turbinates, butachlor causes stomach tumors, and metolachlor causes liver tumors. It has been suggested that the carcinogenicity of these compounds involves a complex metabolic activation pathway leading to a DNA-reactive dialkylbenzoquinone imine. Important intermediates in this pathway are 2-chloro-N-(2,6-diethylphenyl)acetamide (CDEPA) produced from alachlor and butachlor and 2-chloro-N-(2-methyl-6-ethylphenyl)acetamide (CMEPA) produced from acetochlor and metolachlor. Subsequent metabolism of CDEPA and CMEPA produces 2,6-diethylaniline (DEA) and 2-methyl-6-ethylaniline (MEA), which are bioactivated through para-hydroxylation and subsequent oxidation to the proposed carcinogenic product dialkylbenzoquinone imine. The current study extends our earlier studies with alachlor and demonstrates that rat liver microsomes metabolize acetochlor and metolachlor to CMEPA (0.065 nmol/min/mg and 0.0133 nmol/min/mg, respectively), whereas human liver microsomes can metabolize only acetochlor to CMEPA (0.023 nmol/min/mg). Butachlor is metabolized to CDEPA to a much greater extent by rat liver microsomes (0.045 nmol/min/mg) than by human liver microsomes (< 0.001 nmol/min/mg). We have determined that both rat and human livers metabolize both CMEPA to MEA (0.308 nmol/min/mg and 0.541 nmol/min/mg, respectively) and CDEPA to DEA (0.350 nmol/min/mg and 0.841 nmol/min/mg, respectively). We have shown that both rat and human liver microsomes metabolize MEA (0.035 nmol/min/mg and 0.069 nmol/min/mg, respectively) and DEA (0.041 nmol/min/mg and 0.040 nmol/min/mg, respectively). We have also shown that the cytochrome P450 isoforms responsible for human metabolism of acetochlor, butachlor, and metolachlor are CYP3A4 and CYP2B6.


Biochimica et Biophysica Acta | 1996

TOBACCO BUDWORM P-GLYCOPROTEIN : BIOCHEMICAL CHARACTERIZATION AND ITS INVOLVEMENT IN PESTICIDE RESISTANCE

Christine L. Lanning; Robert L. Fine; James J. Corcoran; Hafez M. Ayad; Randy L. Rose; Mohamed B. Abou-Donia

Since pesticides have been shown to interact with P-glycoprotein (P-gp), the purpose of this study was to examine the possible role of P-gp in pesticide resistance in the tobacco budworm (Heliothis virescens). Using three P-gp antibodies, P-gp expression in various resistant populations of tobacco budworms was found to be 2-6-times that of the susceptible larvae. Tobacco budworm P-gp was glycosylated and localized primarily in the cuticle and fat body with little expression in the mid gut. To determine the role of P-gp in pesticide resistance, resistant tobacco budworm larvae were treated with a P-gp inhibitor, quinidine, and challenged with various doses of thiodicarb. Inhibition of P-gp decreased the LD50 for thiodicarb by a factor of 12.5. Quinidine treatment did not result in a significant inhibition of the P-450 system nor did it alter the feeding of the larvae, suggesting the potential involvement of P-gp in pesticide resistance. An age-dependent increase in P-gp expression was detected in resistant larvae as compared to control, susceptible larvae. This correlates with the reported age-dependent increase in resistance and is further evidence supporting the role of P-gp in the development of pesticide resistance.


Pesticide Biochemistry and Physiology | 2002

In vitro human metabolism of permethrin: the role of human alcohol and aldehyde dehydrogenases

Jonghoon Choi; Randy L. Rose; Ernest Hodgson

Permethrin is a pyrethroid insecticide widely used in agriculture and public health. It has been suggested that permethrin may interact with other chemicals used during military deployments and, as a result, be a potential cause of Gulf War Related Illness. To determine the causal relationship between permethrin and human health effects, the basic enzymatic pathway of permethrin metabolism in humans should be understood. In the present study we report that trans-permethrin is metabolized in human liver fractions, producing phenoxybenzyl alcohol (PBOH) and phenoxybenzoic acid (PBCOOH). We identified human alcohol (ADH) and aldehyde dehydrogenases (ALDH) as the enzymes involved in the oxidation of phenoxybenzyl alcohol, the permethrin hydrolysis product, to phenoxybenzoic acid by way of phenoxybenzaldehyde (PBCHO). Cis-permethrin was not significantly metabolized in human liver fractions. Cytochrome P450 isoforms were not involved either in the hydrolysis of trans-permethrin or in the oxidation of PBOH to PBCOOH. Purified ADH isozymes oxidized PBOH to PBCHO and PBOH was a preferred substrate to ethyl alcohol. Purified ALDH was responsible for PBCHO oxidation to PBCOOH with similar substrate affinity to a previously known substrate, benzyl alcohol. Based on these observations, it appears that PBOH is oxidized to PBCHO by ADH and subsequently to PBCOOH by ALDH, although PBCHO does not accumulate during microsomal incubation. In order to analyze permethrin and its metabolites, previous HPLC-UV methods had to be re-validated and modified. The resulting refined HPLC-UV method is described in detail.


Drug Metabolism and Disposition | 2005

IN VITRO METABOLISM OF NAPHTHALENE BY HUMAN LIVER MICROSOMAL CYTOCHROME P450 ENZYMES

Taehyeon M. Cho; Randy L. Rose; Ernest Hodgson

The Polycyclic Aromatic Hydrocarbon Naphthalene Is An Environmental Pollutant, A Component Of Jet Fuel, And, Since 2000, Has Been Reclassified As A Potential Human Carcinogen. Few Studies Of The In Vitro Human Metabolism Of Naphthalene Are Available, And These Focus Primarily On Lung Metabolism. The Current Studies Were Performed To Characterize Naphthalene Metabolism By Human Cytochromes P450. Naphthalene Metabolites From Pooled Human Liver Microsomes (Phlms) Were Trans-1,2-Dihydro-1,2-Naphthalenediol (Dihydrodiol), 1-Naphthol, And 2-Naphthol. Metabolite Production Generated KM Values Of 23, 40, And 116 μM And VMax Values Of 2860, 268, And 22 Pmol/Mg Protein/Min, Respectively. P450 Isoform Screening Of Naphthalene Metabolism Identified Cyp1A2 As The Most Efficient Isoform For Producing Dihydrodiol And 1-Naphthol, And Cyp3A4 As The Most Effective For 2-Naphthol Production. Metabolism Of The Primary Metabolites Of Naphthalene Was Also Studied To Identify Secondary Metabolites. Whereas 2-Naphthol Was Readily Metabolized By Phlms To Produce 2,6- And 1,7-Dihydroxynaphthalene, Dihydrodiol And 1-Naphthol Were Inefficient Substrates For Phlms. A Series Of Human P450 Isoforms Was Used To Further Explore The Metabolism Of Dihydrodiol And 1-Naphthol. 1,4-Naphthoquinone And Four Minor Unknown Metabolites From 1-Naphthol Were Observed, And Cyp1A2 And 2D6*1 Were Identified As The Most Active Isoforms For The Production Of 1,4-Naphthoquinone. Dihydrodiol Was Metabolized By P450 Isoforms To Three Minor Unidentified Metabolites With Cyp3A4 And Cyp2A6 Having The Greatest Activity Toward This Substrate. The Metabolism Of Dihydrodiol By P450 Isoforms Was Lower Than That Of 1-Naphthol. These Studies Identify Primary And Secondary Metabolites Of Naphthalene Produced By Phlms And P450 Isoforms.


Xenobiotica | 1998

Physiological factors affecting protein expression of flavin-containing monooxygenases 1, 3 and 5

Nathan J. Cherrington; Yan Cao; J. W. Cherrington; Randy L. Rose; Ernest Hodgson

1. The mouse and rat exhibit substantial differences in the gender expression of flavin-containing monooxygenase (FMO) forms. Hepatic FMO1 is gender-dependent in both species, selective to the male in rat, female in mouse. Human FMO1 is nearly undetectable. FMO3 in mouse is gender-specific to the female, but gender-independent in rat and man. FMO5 is gender-independent for mouse, rat and man. 2. Gender differences in substrate metabolism do not reflect overall FMO or isoform differences. Methimazole, imipramine and thiobenzamide are much better substrates for FMO1 than for FMO3 or FMO5. 3. Activities of microsomal samples toward these substrates reflect the relative abundance of FMO1. Hepatic samples show a 3-fold greater activity toward methimazole in the female mouse and male rat. Human microsomal samples show minimal activity. 4. Developmentally, FMO1 and FMO5 are expressed in foetuses as early as gestation days 15 and 17 and equally between genders until puberty. FMO3 is not found until 2 weeks post-partum and is found equally in the male and female until 6 weeks post-partum when it becomes undetectable in the male. 5. An event takes place after birth but before puberty that confers the ability to produce FMO3. The developmental pattern observed for mouse FMO3 is similar to human FMO3.


Drug Metabolism and Disposition | 2006

Metabolism of endosulfan-alpha by human liver microsomes and its utility as a simultaneous in vitro probe for CYP2B6 and CYP3A4.

Richard Ct Casabar; Andrew D. Wallace; Ernest Hodgson; Randy L. Rose

Endosulfan-α is metabolized to a single metabolite, endosulfan sulfate, in pooled human liver microsomes (Km = 9.8 μM, Vmax = 178.5 pmol/mg/min). With the use of recombinant cytochrome P450 (P450) isoforms, we identified CYP2B6 (Km = 16.2 μM, Vmax = 11.4 nmol/nmol P450/min) and CYP3A4 (Km = 14.4 μM, Vmax = 1.3 nmol/nmol P450/min) as the primary enzymes catalyzing the metabolism of endosulfan-α, although CYP2B6 had an 8-fold higher intrinsic clearance rate (CLint = 0.70 μl/min/pmol P450) than CYP3A4 (CLint = 0.09 μl/min/pmol P450). Using 16 individual human liver microsomes (HLMs), a strong correlation was observed with endosulfan sulfate formation and S-mephenytoin N-demethylase activity of CYP2B6 (r2 = 0.79), whereas a moderate correlation with testosterone 6 β-hydroxylase activity of CYP3A4 (r2 = 0.54) was observed. Ticlopidine (5 μM), a potent CYP2B6 inhibitor, and ketoconazole (10 μM), a selective CYP3A4 inhibitor, together inhibited approximately 90% of endosulfan-α metabolism in HLMs. Using six HLM samples, the percentage total normalized rate (% TNR) was calculated to estimate the contribution of each P450 in the total metabolism of endosulfan-α. In five of the six HLMs used, the percentage inhibition with ticlopidine and ketoconazole in the same incubation correlated with the combined % TNRs for CYP2B6 and CYP3A4. This study shows that endosulfan-α is metabolized by HLMs to a single metabolite, endosulfan sulfate, and that it has potential use, in combination with inhibitors, as an in vitro probe for CYP2B6 and 3A4 catalytic activities.


Drug Metabolism and Disposition | 2006

Inhibition of the human liver microsomal and human cytochrome P450 1A2 and 3A4 metabolism of estradiol by deployment-related and other chemicals.

Khawja A. Usmani; Taehyeon M. Cho; Randy L. Rose; Ernest Hodgson

Cytochromes P450 (P450s) are major catalysts in the metabolism of xenobiotics and endogenous substrates such as estradiol (E2). It has previously been shown that E2 is predominantly metabolized in humans by CYP1A2 and CYP3A4 with 2-hydroxyestradiol (2-OHE2) the major metabolite. This study examines effects of deployment-related and other chemicals on E2 metabolism by human liver microsomes (HLM) and individual P450 isoforms. Kinetic studies using HLM, CYP3A4, and CYP1A2 showed similar affinities (Km) for E2 with respect to 2-OHE2 production. Vmax and CLint values for HLM are 0.32 nmol/min/mg protein and 7.5 μl/min/mg protein; those for CYP3A4 are 6.9 nmol/min/nmol P450 and 291 μl/min/nmol P450; and those for CYP1A2 are 17.4 nmol/min/nmol P450 and 633 μl/min/nmol P450. Phenotyped HLM use showed that individuals with high levels of CYP1A2 and CYP3A4 have the greatest potential to metabolize E2. Preincubation of HLM with a variety of chemicals, including those used in military deployments, resulted in varying levels of inhibition of E2 metabolism. The greatest inhibition was observed with organophosphorus compounds, including chlorpyrifos and fonofos, with up to 80% inhibition for 2-OHE2 production. Carbaryl, a carbamate pesticide, and naphthalene, a jet fuel component, inhibited ca. 40% of E2 metabolism. Preincubation of CYP1A2 with chlorpyrifos, fonofos, carbaryl, or naphthalene resulted in 96, 59, 84, and 87% inhibition of E2 metabolism, respectively. Preincubation of CYP3A4 with chlorpyrifos, fonofos, deltamethrin, or permethrin resulted in 94, 87, 58, and 37% inhibition of E2 metabolism. Chlorpyrifos inhibition of E2 metabolism is shown to be irreversible.


Chemico-Biological Interactions | 1999

In vitro metabolism of alachlor by human liver microsomes and human cytochrome P450 isoforms

Scott Coleman; Siming Liu; Russell J. Linderman; Ernest Hodgson; Randy L. Rose

Alachlor (2-chloro-N-methoxymethyl-N-(2,6-diethylphenyl)acetamide) is a widely used pre-emergent chloroacetanilide herbicide which has been classified by the USEPA as a probable human carcinogen. The putative carcinogenic metabolite, 2,6-diethylbenzoquinone imine (DEBQI), is formed through a complex series of oxidative and non-oxidative steps which have been characterized in rats, mice, and monkeys but not in humans. A key metabolite leading to the formation of DEBQI is 2-chloro-N-(2,6-diethylphenyl)acetamide (CDEPA). This study demonstrates that male human liver microsomes are able to metabolize alachlor to CDEPA. The rate of CDEPA formation for human liver microsomes (0.0031 +/- 0.0007 nmol/min per mg) is significantly less than the rates of CDEPA formation for rat liver microsomes (0.0353+/-0.0036 nmol/min per mg) or mouse liver microsomes (0.0106 +/- 0.0007). Further, we have screened human cytochrome P450 isoforms 1A1, 1A2, 2B6, 2C8, 2C9, 2C18, 2C19, 2D6, 2E1, and 3A4 and determined that human CYP 3A4 is responsible for metabolism of alachlor to CDEPA. Further work is necessary to determine the extent to which humans are able to metabolize CDEPA through subsequent metabolic steps leading to the formation of DEBQI.


Toxicology Letters | 1995

Pesticide-metabolizing enzymes.

Ernest Hodgson; Randy L. Rose; Doug-Young Ryu; G. Falls; Bonnie L. Blake; Patricia E. Levi

Pesticides are known to function as substrates, inhibitors and inducers of drug-metabolizing enzymes, with the same compound frequently acting in more than one of these roles. Current studies of phase I metabolism of pesticides include cytochrome P450 (P450) and the flavin-containing monooxygenase (FMO), with particular reference to individual isozymes. In mouse liver, the level of FMO1 is gender dependent, FMO3 is gender specific, while FMO5 appears to be gender independent. The isozyme specificity of methylenedioxyphenyl synergists for induction of P450 in mouse liver involves P450s 1A1, 1A2 and 2B10, including a non-Ah receptor-dependent mechanism for 1A2 induction. The substrate specificity of mouse and human P450 and FMO isozymes is discussed.

Collaboration


Dive into the Randy L. Rose's collaboration.

Top Co-Authors

Avatar

Ernest Hodgson

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Yan Cao

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Patricia E. Levi

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Jun Tang

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Parikshit C. Das

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

R. M. Roe

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Andrew D. Wallace

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Khawja A. Usmani

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

R. Michael Roe

North Carolina State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge