Network


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

Hotspot


Dive into the research topics where Thomas W. Jones is active.

Publication


Featured researches published by Thomas W. Jones.


Toxicology and Applied Pharmacology | 1990

Renal cysteine conjugate β-lyase-mediated toxicity studied with primary cultures of human proximal tubular cells

John C. Chen; James L. Stevens; Anna L. Trifillis; Thomas W. Jones

The beta-lyase pathway has been shown to mediate the nephrotoxicity of S-cysteine conjugates of a variety of haloalkenes in a number of animal models in vitro and in vivo. However, there is no information available concerning this mechanism of bioactivation in human tissues. In this investigation a well-characterized model of human proximal tubule epithelial cells, the presumed target cell, was used to investigate the toxicity of a series of glutathione and cysteine conjugates of nephrotoxic haloalkenes. Both S-(1,2-dichlorovinyl)-glutathione (DCVG) and S-(1,2-dichlorovinyl)-L-cysteine (DCVC) caused dose-dependent toxicity over a range of 25 to 500 microM. DCVC was consistently found to be more toxic than DCVG, but the inclusion of gamma-glutamyltransferase (0.5 U/ml) increased the toxicity of DCVG to that observed with an equimolar concentration of DCVC, indicating that metabolism to the cysteine conjugate is an important rate-limiting step in this in vitro model. S-(1,2,3,4,4-Pentachlorobutadienyl)-L-cysteine, S-(2-chloro-1,1,2-trifluoroethyl)-L-cysteine, and S-(1,1,2,2-tetrafluoroethyl)-L-cysteine were also found to be toxic to human proximal tubular cells. Incubation with [35S]DCVC resulted in covalent binding of 35S-label, which increased linearly to a final level of 1.05 nmol/mg protein at 6 hr. Aminooxyacetic acid (250 microM), an inhibitor of pyridoxal phosphate-dependent enzymes such as beta-lyase, protected the cells from the toxicity of all of the cysteine conjugates and inhibited the covalent binding of 35S-label from [35S]DCVC to cellular macromolecules. The results of the present study provide the first evidence that human proximal tubular cells are sensitive to the toxicity of glutathione and/or cysteine conjugates of a variety of chloro- and fluoroalkenes which are activated via the beta-lyase pathway. The implications for human health are discussed.


Toxicology and Applied Pharmacology | 1990

Differences in the localization and extent of the renal proximal tubular necrosis caused by mercapturic acid and glutathione conjugates of 1,4-naphthoquinone and menadione☆

Serrine S. Lau; Thomas W. Jones; Robert J. Highet; Barbara A. Hill; Terrence J. Monks

We have previously demonstrated that administration of various benzoquinol-glutathione (GSH) conjugates to rats causes renal proximal tubular necrosis and the initial lesion appears to lie within that portion of the S3 segment within the outer stripe of the outer medulla (OSOM). The toxicity may be a consequence of oxidation of the quinol conjugate to the quinone followed by covalent binding to tissue macromolecules. We have therefore synthesized the GSH and N-acetylcysteine conjugates of 2-methyl-1,4-naphthoquinone (menadione) and 1,4-naphthoquinone. The resulting conjugates have certain similarities to the benzoquinol-GSH conjugates, but the main difference is that reaction with the thiol yields a conjugate which remains in the quinone form. 2-Methyl-3-(N-acetylcystein-S-yl)-1,4-naphthoquinone caused a dose-dependent (50-200 mumol/kg) necrosis of the proximal tubular epithelium. The lesion involved the terminal portion of the S2 segment and the S3 segment within the medullary ray. At the lower doses, that portion of the S3 segment in the outer stripe of the outer medulla displayed no evidence of necrosis. In contrast, 2-methyl-3-(glutathion-S-yl)-1,4-naphthoquinone (200 mumol/kg) caused no apparent histological alterations to the kidney. 2-(Glutathion-S-yl)-1,4-naphthoquinone and 2,3-(diglutathion-S-yl)-1,4-naphthoquinone (200 mumol/kg) were relatively weak proximal tubular toxicants and the lesion involved the S3 segment at the junction of the medullary ray and the OSOM. A possible reason(s) for the striking difference in the toxicity of the N-acetylcysteine conjugate of menadione, as opposed to the lack of toxicity of the GSH conjugate of menadione, is discussed. The basis for the localization of the lesion caused by 2-methyl-3-(N-acetylcystein-S-yl)-1,4-naphthoquinone requires further study.


Toxicology and Applied Pharmacology | 1991

Inhibition of γ-glutamyl transpeptidase potentiates the nephrotoxicity of glutathione-conjugated chlorohydroquinones☆

J.J.W.M. Mertens; J.H.M. Temmink; Peter J. van Bladeren; Thomas W. Jones; Herng Hsiang Lo; Serrine S. Lau; Terrence J. Monks

Administration of either 2,5-dichloro-3-(glutathion-S-yl)-1, 4-benzoquinone (DC-[GSyl]BQ) or 2,5,6-trichloro-3-(glutathion-S-yl)-1,4-benzoquinone (TC-[GSyl]BQ) to male Sprague-Dawley rats caused dose-dependent (50-200 mumol/kg; iv) renal proximal tubular necrosis, as evidenced by elevations in blood urea nitrogen (BUN), and in the urinary excretion of lactate dehydrogenase (LDH), gamma-glutamyl transpeptidase (gamma-GT) and glucose. Renal proximal tubular necrosis was also confirmed by histological examination of kidney slices prepared from DC-(GSyl)BQ- and TC-(GSyl)BQ-treated animals. Administration of the corresponding hydroquinone conjugates (DC-[GSyl]HQ and TC-[GSyl]HQ), prepared by reducing the quinones with a threefold molar excess of ascorbic acid, resulted in a substantial increase in nephrotoxicity. Moreover, in contrast to other glutathione (GSH)-conjugated hydroquinones, the nephrotoxicity of both DC-(GSyl)HQ and TC-(GSyl)HQ was potentiated when rats were pretreated with AT-125, an irreversible inhibitor of gamma-GT. Neither the quinone-GSH nor the hydroquinone-GSH conjugates caused any effect on liver histology or serum glutamate-pyruvate transaminase levels. The results suggest that coadministration of ascorbic acid with DC-(GSyl)BQ or TC-(GSyl)BQ decreases their interactions with extrarenal nucleophiles, including plasma proteins, and thus increases the concentration of the conjugates delivered to the kidney, and hence toxicity. Furthermore the ability of AT-125 to potentiate the nephrotoxicity of DC-(GSyl)HQ and TC-(GSyl)HQ suggests that metabolism of these conjugates by gamma-GT constitutes a detoxication reaction.


Toxicology and Applied Pharmacology | 1988

Stimulation of the renal endoplasmic reticulum calcium pump: a possible biomarker for platinate toxicity.

Linda M. De Witt; Thomas W. Jones; Leon Moore

Antitumor platinum compounds such as cisplatin are frequently nephrotoxic. The mechanism of nephrotoxicity has not been determined. It has been proposed that some toxicants may act by interfering with the mechanisms that control cellular Ca2+ homeostasis. An important factor in the regulation of cytosolic Ca2+ is the endoplasmic reticulum (ER) calcium pump. The activity of this pump was determined by measuring ATP-dependent microsomal sequestration of 45Ca. Administration of nephrotoxic doses of platinum compounds to rats was associated with an increase in renal ER calcium pump activity. This was the earliest response observed after cisplatin treatment (it occurred within 4 hr) and preceded increases in blood urea nitrogen and serum creatinine by at least 1 day. The dose-response curve for the increase in renal ER calcium pump activity was similar to the increase in the number and size of smooth ER aggregates observed in the S3 segment of the proximal tubule 24 hr following cisplatin administration. Only minor morphological changes were observed at this time. There was a significant increase in calcium content of kidneys of rats 24 hr after treatment with a dose of cisplatin that caused a maximal increase in ER calcium pump activity. This indicates that a disruption of normal calcium homeostasis may occur before histological evidence of nephrotoxicity. Platinates that were not toxic to the kidney did not elevate renal ER calcium pump activity. It is suggested that the activity of the ER calcium pump may be a useful biomarker for cellular toxicity and may be a factor in the mechanism of toxicity.


Chemico-Biological Interactions | 1988

Features of microsomal and cytosolic glutathione conjugation of hexachlorobutadiene in rat liver.

Alf Wallin; Robert G. Gerdes; Ralf Morgenstern; Thomas W. Jones; Kari Ormstad

Hepatic GSH conjugation is the initial step in the mammalian biotransformation of hexachloro-1,3-butadiene (HCBD) and analogous haloalkenes. The present paper reports an in vitro investigation of the glutathione-dependent conversion of HCBD to water-soluble products, i.e. the enzyme-catalyzed conjugation of HCBD with GSH. The method employed avoids artifacts due to the volatility, low solubility and hydrophobic nature of the chloro-carbon substrate. In order to assess the relative importance of membrane-bound and cytosolic glutathione S-transferase in the conjugation process, microsomal and cytosolic fractions from adult rat liver were tested separately for their ability to promote water solubilisation of the substrate. In addition, microsomal purified and liposomally reconstituted glutathione S-transferase, were tested. The reaction exhibited Michaelis-Menten kinetics, and conjugation rates were linear for at least 20 min. The hepatic microsomal fraction metabolized HCBD 116 times faster than the cytosolic fraction when substrate saturated. Both mono- and bis-substituted conjugates were formed by microsomal as well as by the cytosolic fraction. Treatment of animals with inducers and the use of specific inhibitors indicated absence of cytochrome P-450 involvement in the formation of water soluble HCBD metabolites and supported the view that microsomal glutathione S-transferase is more important in catalyzing GSH conjugation of this haloalkene than the cytosolic forms of transferases.


Toxicology | 1990

Species differences in renal γ-glutamyl transpeptidase activity do not correlate with susceptibility to 2-bromo-(diglutathion-S-yl)-hydroquinone nephrotoxicity

Serrine S. Lau; Thomas W. Jones; Rowena Sioco; Barbara A. Hill; Roger K. Pinon; Terrence J. Monks

Administration of 2-bromo-(diglutathion-S-yl)hydroquinone (2-Br-[diGSyl]HQ) (10-30 mumol/kg; i.v.) to rats causes severe renal proximal tubular necrosis. gamma-Glutamyl transpeptidase (gamma-GT) catalyses the first step in the metabolism of glutathione (GSH) and its S-conjugates and the toxicity of 2-Br-(diGSyl)HQ can be emeliorated by inhibition of renal gamma-GT. Species differences in the specific activity of renal gamma-GT have been reported and we now describe the relationship between renal gamma-GT and species differences in susceptibility to 2-Br-(diGSyl)HQ nephrotoxicity. Although rats exhibited the highest specific activity of renal gamma-GT, and were the most sensitive species toward 2-Br-(diGSyl)HQ-mediated nephrotoxicity, renal gamma-GT activity did not correlate with susceptibility in the other species examined. Indeed, the guinea pig, which expressed the lowest activity of renal gamma-GT between the species (8% of the rat) was the only other rodent found to be responsive toward 2-Br-(diGSyl)HQ at the highest dose tested (200 mumol/kg; intracardiac). Thus, factors other than gamma-GT activity probably play an important role in modulating species susceptibility to 2-Br-(diGSyl)HQ nephrotoxicity. Although the reason(s) for the interspecies variation in response to 2-Br-(diGSyl)HQ are unclear at present, it seems possible that differences in both renal biochemistry, such as differences in the relative activities of cysteine conjugate N-acetyl transferase and deacetylase, and renal physiology, contribute to the observed results.


Toxicology and Applied Pharmacology | 1991

Nephrotoxicity of 2-bromo-(cystein-s-yl) hydroquinone and 2-bromo-(N-acetyl-l-cystein-S-yl) hydroquinone thioethers☆☆☆

Terrence J. Monks; Thomas W. Jones; Barbara A. Hill; Serrine S. Lau

The in vivo toxicity of isomeric cystein-S-yl and N-acetylcystein-S-yl conjugates of 2-bromohydroquinone was determined in male Sprague-Dawley rats. 2-Bromo-(dicystein-S-yl)hydroquinone [2-Br-(diCYS)HQ] and 2-bromo-(di-N-acetyl-L-cystein-S-yl)hydroquinone [2-Br-(diNAC)HQ] were considerably more nephrotoxic than their corresponding monosubstituted thioethers and 2-Br-(diCYS)HQ was more nephrotoxic than 2-Br-(diNAC)HQ. 2-Br-(diCYS)HQ caused elevations in blood urea nitrogen (BUN) concentrations and increases in the urinary excretion of glucose, lactate dehydrogenase (LDH), and gamma-glutamyl transpeptidase (gamma-GT) at a dose of 25 mumol/kg (iv). In contrast, 2-Br-(diNAC)HQ caused significant elevations in BUN at 100 mumol/kg and glucosuria and enzymuria at 50 mumol/kg. 2-Br-3-(CYS)HQ and 2-Br-5&6-(CYS)HQ caused increases in the biochemical indices of nephrotoxicity at doses between 50 and 150 mumol/kg whereas 2-Br-5-(NAC)HQ and 2-Br-6-(NAC)HQ required doses of 150-200 mumol/kg to cause smaller, though significant increases in urinary glucose, gamma-GT, and LDH excretion. The histological alterations caused by each thioether were qualitatively similar; only differences in the extent of the renal proximal tubular damage were observed. The initial lesion appears to involve the cells of the medullary ray and the S3M within the outer stripe of the outer medulla. The in vivo nephrotoxicity of 2-Br-(DiCYS)HQ, 2-Br-(diNAC)HQ, and the most potent monosubstituted thioethers, 2-Br-5&6-(CYS)HQ and 2-Br-6-(NAC)HQ, was investigated further. Pretreatment of animals with aminooxyacetic acid, an inhibitor of cysteine conjugate beta-lyase (beta-lyase), had no effect on the toxicity of 2-Br-(diCYS)HQ, partially inhibited the toxicity of 2-Br-5&6-(CYS)HQ, and almost completely protected against the toxicity of both 2-Br-6-(NAC)HQ and 2-Br-(diNAC)HQ. Thus, the nephrotoxicity of 2-Br-5&6-(CYS)HQ, 2-Br-6-(NAC)HQ, and 2-Br-(diNAC)HQ may be mediated, in part, via their processing by beta-lyase. Pretreatment of animals with probenecid, an inhibitor of renal organic anion transport, completely protected against the toxicity of 2-Br-(diNAC)HQ but had no effect on the toxicity of the other thioethers.


Toxicologic Pathology | 1985

Cis-Diamminedichloroplatinum (II)-induced Acute Renal Failure in the Rat: Enzyme Histochemical Studies

Thomas W. Jones; Shreekant Chopra; James S. Kaufman; Walter Flamenbaum; Benjamin F. Trump

Enzyme histochemical techniques were utilized to examine the progression and extent of proximal tubular injury during the development of cis-diamminedichloroplatinum (II) (CDDP)-induced acute renal failure. Acute renal failure was induced in male rats by the intraperitoneal administration of 10 mg CDDP/kg body weight. At 6, 24, 48, 72, and 96 hr following treatment, renal function was assessed and tissue was collected for renal morphologic and enzyme histochemical studies. The enzymes examined were γ-glutamyl transpeptidase, alkaline phosphatase, sodium-potassium ATPase (nitrophenyl phosphatase), acid phosphatase, glucose-6-phosphatase, succinic dehydrogenase, α-glycerophosphate dehydrogenase, and lactic dehydrogenase. By 24 hr, the activity of acid phosphatase was reduced throughout the proximal tubule, with the greatest decrease occurring in the P3 segment of the proximal tubule located in the outer stripe of the outer medulla. Changes in the histochemical staining of the remaining enzymes were not consistently observed until 48 or, in some cases, 72 hr. These alterations involved all portions of the proximal tubule with the most severe changes involving P3. The results of the enzyme histochemical studies along with the morphologic findings indicating that the initiation of CDDP-induced acute renal failure, first apparent at 48 hr in this model, is associated with cell injury throughout the proximal tubule. The majority of the histochemical changes did not become apparent until late in the course of tubular injury. This suggests that most of the changes in enzyme activity represent nonspecific effects of CDDP-induced tubular injury, as opposed to direct enzyme inhibition by the drug.


Toxicology Letters | 1990

The role of damage and proliferation in renal carcinogenesis

James L. Stevens; Thomas W. Jones

Many carcinogens and/or toxins cause tissue damage. Wounding also increases the frequency of transformation and complements the formation of tumors [l-5]. With many carcinogens, it is difficult to dissociate the appearance of tumors from tissue damage; some compounds are carcinogens only at toxic doses [6]. The hypothesis that ‘epigenetic’ or ‘non-genotoxic’ events play a role in transformation arose in part as an attempt to explain the role of wounding in cancer. The important question is: How does wounding promote or lead directly to cell transformation? A number of hypotheses and observations draw a parallel between transformation and wound healing. Bryan [l] reviewed the early work on wounding and carcinogenesis and suggested that cancer cells resemble an exaggerated ‘reaction to injury’, particularly with respect to tissue renewal. Pierce [7] proposed that cancers ‘ . . . are a caricature of tissue renewal [with] a marked capacity to proliferate and a limited capacity to differentiate.’ Potter’s ‘blocked ontogeny’ hypothesis [8] states that differentiation of cancer cells is arrested in states resembling stages of the normal developmental process. Dvorak’s [9] elegant studies on solid tumor stroma led him to the conclusion that tumors are ‘wounds that will not heal’. Therefore, cancer cells may have similarity with tissue renewal during the wound healing. Wounding may play several roles in carcinogenesis by: (1) providing a provisional


Toxicologic Pathology | 2017

Regulatory Forum Opinion Piece*: Use and Utility of Animal Models of Disease for Nonclinical Safety Assessment: A Pharmaceutical Industry Survey

Sherry J. Morgan; Jessica Couch; Peggy Guzzie-Peck; Douglas A. Keller; Ray Kemper; Monicah A. Otieno; Robert J. Schulingkamp; Thomas W. Jones

An Innovation and Quality (IQ) Consortium focus group conducted a cross-company survey to evaluate current practices and perceptions around the use of animal models of disease (AMDs) in nonclinical safety assessment of molecules in clinical development. The IQ Consortium group is an organization of pharmaceutical and biotechnology companies with the mission of advancing science and technology. The survey queried the utilization of AMDs during drug discovery in which drug candidates are evaluated in efficacy models and limited short-duration non-Good Laboratory Practices (GLP) toxicology testing and during drug development in which drug candidates are evaluated in GLP toxicology studies. The survey determined that the majority of companies used AMDs during drug discovery primarily as a means for proactively assessing potential nonclinical safety issues prior to the conduct of toxicology studies, followed closely by the use of AMDs to better understand toxicities associated with exaggerated pharmacology in traditional toxicology models or to derisk issues when the target is only expressed in the disease state. In contrast, the survey results indicated that the use of AMDs in development is infrequent, being used primarily to investigate nonclinical safety issues associated with targets expressed only in disease states and/or in response to requests from global regulatory authorities.

Collaboration


Dive into the Thomas W. Jones's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Barbara A. Hill

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James L. Stevens

W. Alton Jones Cell Science Center

View shared research outputs
Top Co-Authors

Avatar

James S. Kaufman

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Walter Flamenbaum

United States Department of Veterans Affairs

View shared research outputs
Top Co-Authors

Avatar

Robert J. Hamburger

United States Department of Veterans Affairs

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge