THESIS "A'5.E.P5%. Ll; to 4"?“ 8‘! w Jl‘tfilg'.’ 1:5 “unfit-i tiiifigliil‘ This is to certify that the dissertation entitled DYNAMICS OF GLUTATHIONE REGULATION IN SCHISTOSOMA MANSONI: CORRELATIONS WITH THE ACUTE EFFECTS OF OLTIPRAZ presented by Dan Douglas Morrison has been accepted towards fulfillment of the requirements for Ph.D. degree in Pharmacology and Tox1cology Major professor \ Date 10/25/84 MS U i: an Afi'mnan'w Action/Equal Opportunity Institution 0-12771 MSU RETURNING MATERIALS: Piace in book drop to uaaAmss remove this checkout from ”- your record. FINES win be charged if book is returned after the date stamped below. DYNAMICS OF GLUTATHIONE REGULATION IN SCHISTOSOMA MANSONI: CORRELATIONS WITH THE ACUTE EFFECTS OF OLTIPRAZ By Dan Douglas Morrison A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Pharmacology and Tbxicology 1984 ABSTRACT DYNAMICS OF GLUTATHIONE REGULATION IN SCHISTOSOMA MANSONI: CORRELATIONS RUTH THE ACUTE EFFECTS OF OUTIPRAZ By Dan Douglas Morrison Glutathione is present in adult Schistosoma mansoni (6.336 :_fl.012 nmoles/mg protein) at significantly lower levels than uninfected host liver tissues (1.051 :_@.013 nmoles/mg protein) or host kidney tissue (6.627 1. 6.613 nmoles/mg protein). Host hepatic glutathione levels decline significantly during the course of an infection by the parasite, while renal cortical glutathione levels are unaffected. Of the enzymes regulating glutathione utilization, glutathione reductase in the male parasite exhibits a specific activity of 10.3 :_4.2 mU/mg protein, 15% that of uninfected liver. The apparent glutathione S-transferase activity depended on substrate tested, 26 :_7 umole conjugate fermed/min/mg protein with p— nitrobenzyl chloride as substrate (13% of hepatic values) and 526 :- 18 pmole conjugate formed/min/mg protein with l-chloro—Z,4-dinitrobenzene as substrate (43% of hepatic values). Male schistosomes exhibited negligible glutathione peroxidase activity (5 :_ 9 nmoles NADPH oxidized/min/mg protein), while the uninfected host exhibited values of 258 :.19 nmoles NADPH oxidized/min/mg protein fbr this enzyme. During the course of infection, the specific activity of hepatic glutathione S-transferase was depressed to 26% of uninfected control values (p— nitrobenzyl chloride as substrate). Similarly, hepatic glutathione peroxidase activity was reduced to 37% of control values. Hepatic glutathione reductase was not significantly affected during the infection. Also, neither of the renal enzymes, glutathione peroxidase or glutathione reductase, were affected. Oltipraz, an antischistosomal compound, effected a significant depletion of parasite and host glutathione levels within 1 h of exposure _i_n y_i__v_o_ and _i_n_ _v_i_t_;_r_g at a dosage of 256 'mg/kg and 10 H5! respectively. Host tissue glutathione levels returned to, or above, control levels by 6 h after oltipraz administration, while parasite glutathione levels remained significantly depressed. Uptake of [35$]cysteine or [3551cystine by schistosomes was inhibited by oltipraz. However, the drug did not alter the relative distribution of label once incorporated into the parasite, indicating that the enzymes of glutathione synthesis were not directly inhibited. Oltipraz significantly depressed surface electrical activity and the tegument potential of male schistosomes. However, both the biochemical and physiological effects of oltipraz (10 H!) were prevented by coincubat— ing the parasite in 1 WE. cysteine, methionine or glutathione. The amelioration of oltipraz-induced effects was not dependent on free thiol groups, as neither dithiothreitol nor Zemercaptoethanol attenuated the schistosomicidal effect of oltipraz. The inactive analog of oltipraz (RP 36 642, 106 p_M_) did not produce effects mimick- ing those seen with oltipraz. ACKNOWLEDGEMENTS I would like to acknowledge the generous support and guidance of my advisor, Dr. James L. Bennett. Dr. Bennett's patience and under- standing, in the genesis and development of this thesis, have helped enable its completion despite some unexpected trials and tribulations during my graduate career, which have given the catchwords 'in sickness and in health' new meaning. My special thanks to Dr. David P. Thompson for perusal of manuscripts (ad nauseum) and for his unique style of ”guillotine“ humor . I am indebted to Drs. 'Iheodore Brody, Ralph Fax and Paul Sato for guidance and assistance with the developnent of the thesis proposal. Ms. H. Cirrito and Ms. I. Mao provided invaluable technical assistance with tissue preparations. Financial support from NIH grants AI19889 and M14993 is gratefully acknowledged. ii TABIEOFCONTENTS PAGE LIST OF TABLES vi LIST OF FIGURES vii LIST OF ABBREVIATIONS ix INTRODUCTION] A. The parasite, Schistosoma mansoni l B. Chemotherapy 2 1. General 2. Oltipraz and structure-activity relationships 3. Oltipraz spectrun of action 4. Oltipraz mechanism of action C. Glutathione and its enzymatic regulation 6 l. Glutathione 2. Enzymatic synthesis 3. Glutathione utilization RATIONAIE 14 iii TABLE OF CCNI'ENTS (con't) MATERIALS AND METHODS A. Preparation of host and parasite tissue B. Glutathione assay C. Manipulation of schistosome glutathione levels in vitro D. High Pressure Liquid Chromatography (HPDC) -—--—-- E. Enzyme assays F. Mechanical, surface electrial and tegumental potential measurements Chapter 1: Best—parasite differences in the regulation of glutathione utilization Introduction Results Discussion Chapter 2: Acute effects of oltipraz and its antagonism .ig vitro Introduction Results iv 15 l6 l6 l7 17 19 20 21 24 29 29 TABLE OF CCNI'ENTS (con't) Discussion 38 Chapter 3: Correlation of schistosomal glutathione levels with physiological parameters Introduction 46 Results 46 Discussion 49 Chapter 4: Effects of schistosomal infection on host regulation of glutathione levels Introduction 55 Results 56 Discussion 59 SUMMARY AND CONCLUSIONS 6% BIBLIOGRAPHY 62 Table mansoni and uninfected mouse liver tissue LIST OF TABLES Drugs used in the treatment of human schistosomiasis Glutathione S-transferase activities of Schistosoma Mouse hepatic glutathione-related enzyme activity: Effect of Schistosoma mansoni infection ‘vi Page 23 58 Figure 10 ll 12 13 LIST OF FIGURES Chemical structure of oltipraz Chemical structure of glutathione Y-glutamyl cycle Chemical structure of buthionine sulfOximine Dose—dependent depletion of schistosomal glutathione by’diamide Oltipraz effect on glutathione content in infected mouse hepatic and renal cortical tissue and in male schistosomes Dose-dependent depletion of schistosomal glutathione by oltipraz Effect of oltipraz and various compounds on schistosome glutathione levels_in vitro Oltipraz effect on schistosomal glutathione levels _in vitro: Aerobic vs anaerobic environment [358]cysteine uptake and incorporation into schistosomes in vitro [14C]glucose uptake into schistosomes in vitro Surface electrical activity of male schistosomes in vivo: Oltipraz effect Tegument potential of male schistosomes in_vitro: Oltipraz effect and antagonism by cysteine 'vii Page 11 22 30 31 32 34 35 36 37 39 Figure 14 15 l6 17 18 19 20 LIST or FIGURES (con't) Hypothetical reaction sequence to explain oxygen- dependency of oltipraz-induced depletion of schistosomal GSH in vitro Dose-dependent depletion of schistosomal glutathione by BCNU Tegument potential of male schistosomes in vitro: Effect of BCNU and antagonism by cysteine Dose-dependent depletion of schistosomal glutathione by 880 Oltipraz effect on schistosome fecundity in vitro —- Effect of various reagents on oltipraz inhibition of schistosome fecundity in vitro Glutathione levels in host hepatic and renal cortical tissues during the course of Schistosoma mansoni infection viii Page 42 47 48 50 51 52 57 AT-125 BSO GRed GSH GTP HPLC HS/RPMI NAD(P)H SCE. LIST OF ABBREVIATIONS Irwas,SS)-a-Amino-3-chloro—4,5—dihydro—S-isoxazoleacetic acid 1,3—bis [2-chloroethyl]-l-nitrosourea L—buthionine-S-R—sulfoximine Dithiothreitol Glutathione peroxidase (EC 1.11.1.9) Glutathione reductase (EC 1.6.4.2) Glutathione, reduced fOrm (Y-L—glutamyl-L—cysteinylglycine) Glutathione transpeptidase (EC 2.3.2.2) Glutathione S—transferase (EC 2.4.1.18) High pressure liquid chromatography 1:1 mixture of horse serum (heat-inactivated) and RPMI 1640, to which penicillin (100 U/ml), streptomycin (100 ug/ml) and Fungizone (0.5 pg/ml) were added Nicotinamide—adenine dinucleotide (phosphate) Standard error of the mean Trichloroacetic acid ix INTRODUCTTON A. The parasite, Schistosoma mansoni: Schistosoma mansoni is a digenetic parasite, which debilitates millions of people in tropical regions throughout the world. The regions of endemic schistosomiasis are restricted to tropical climates because of the presence of the snail vector in which the schistosome develops during one stage of its two-host life cycle. '§;_ mansoni adults, residing in the mesentaric venules lining the intestines of the human, oviposit approximately 300 eggs/day/worm pair [99]. Up to 70% of these are dislodged from the venule into the blood stream and become entrapped within the hepatic sinusoids of the liver or the microfenestrations of the spleen, resulting in the hepatosplenic pathology which characterizes long-term, heavy worm burden infections [130,131,132]. The overt disease symptoms, hepatomegaly, splenomegaly, portal hypertension and esophageal varices, can be prevented, even in mice heavily infected with schistosomes by inhibition of oviposition. Also, the disease state shows signs of amelioration, by gradual host repair of damaged tissues, if egg production is halted after the onset of hepatosplenic disease [129]. An increase in the prevalence and intensity of schistosomiasis is occurring in the tropics because agricultural irrigation is rapidly expanding in countries where the disease is endemic, providing a larger habitat fer the snail host. To interrupt transmission of the disease requires effective sanitary waste disposal, large—scale snail control, or mass chemotherapy. Hewever, field-trials have indicated that the best results are obtained with chemotherapy [78], and chemotherapy will likely remain the only viable approach in the control of the disease for the extended future. HOwever, due to the proclivity of the human population to become reinfected, mass chemotherapy is an expensive temporary solution. B. Chemotherapy: 1. General: Several compounds are available which exhibit antischistosomal activity (Table 1, from [16]), however, the use of the antimonials, niridazole and hycanthone is limited due to host toxicity. Metrifonate is not effective in eliminating mansoni schistosomiasis, which limits its use. Research is warranted on the other antischistosomal compounds to attempt to determine their modes of action in efforts to design more efficacious drugs or cheaper substitutes that still retain schistosomicidal activity. Further research is also warranted to forestall or circunvent possible problems of resistance that may develop to drugs currently available and in use, or the possible discovery of untoward effects of the newer drugs on the host. Ideally, the antiparasitic would be effective in a single oral dose, of low cost, and effective against all stages of the parasite Table 1: Drugs Used in the Treatment of Human Schistosomiasis Antischistosomal Antischistosomal Cbmments compoung Spectrum of (date) Action Antimonials __._ _._, __._ j_._, _S__._ __:_ Limited use due (1918) to toxicity Metrifonate _;__;_ Minor side (1962) effects Niridazole EL.EEJ.§_._; Cbnvulsant, (1965) carcinogenic Hycanthone §;.EkJ _J__;_ Hepatic necrosis, (1967) carcinogenic Oxamniquine §;.Eh. Minor side (1973) effects Amoscanate §;_ . Hepatotoxicity (1978) _S_:__r_n_._ (?) ,_S_.__h_._ (7) Praziquantel _S_._ m., .5; ., _S__.__h__ (1979) Oltipraz §.:. _I_n_:_, _S_._ ___:_ (1979) a Date of first reported use in humans with schistosomiasis. Schistosoma mansoni (S. m.), Schistosoma haematobium (S. h.) and Sohi§tosoma japonicum (S. j.). [67] . 2. Oltipraz structure and structure-activity relationships: Oltipraz (35 972 R.P.; 4-methy1—5-[2-pyrizinyl]-l,2—dithiol-3-thione; Figure l) was synthesized and patented in the mid-1970's by Rhone Poulenc Sante, Paris [9]. Congeners of this drug exhibit amebicidal [9] and fungicidal [12] activity, while some other compounds with the 1,2-dithiole-3-thione ring structure exhibit diuretic activity [38]. The structure-activity relationship for oltipraz is rather stringent [25]. The presence of the thione and an adjacent aromatic ring con- taining two nitrogens are structural properties required for antischistosomal activity [23]. Further, while oltipraz is extensively metabolized [19], none of the metabolites which have been identified possess any antischistosomal activity [77]. 3. Oltipraz spectrum _o__f action: In man, oltipraz is effective in erradicating _S_.mansoni, _S_.haematobium and _S_.intercalatum infections [49,108] , but the drug is ineffective against _S_. jamnicum [24]. Oltipraz is less active against schistosomula than against mature adult schistosomes [89] . 4. Oltipraz mechanism 313' action: Oltipraz is a slow-acting drug, two or more months may pass before the worm burden is totally erradicated with a curative dose. In mice given very high doses of the drug, evidence of worm lethality is observed after 48 h, with all schistosomes being killed by 14 days post-treatment [89] . The mechanism of action which leads eventually to the schistosomacidal Figure 1: Chemical structure of oltipraz. OLTIPRAZ outcome remains unknown. There are no published reports on acute effects of this drug, nor have reports been published 0“.12”21E£9 studies to determine the direct action of this compound on the parasite which are independent of the variables of the host response to drug action. With oltipraz, and various structural analogs of oltipraz, a positive correlation exists between antischistosomal activity and the depression of glutathione levels in the parasite [25]. Although not studied acutely, these investigators also observed a concomittant elevation in the glutathione levels of various host tissues post-treatment, suggesting that a biochemical difference exists between host and parasite in the regulation of this important intracellular thiol compound. C. Glutathione and enzymatic regulation of glutathione: l. Glutathione: Glutathione (Y-L—glutamyl-L-cysteinylglycine; GSH) is a tripeptide of molecular weight 307.33, shown in Figure 2. The pKa of the sulfhydryl is 9.2, therefbre GSH carries a net negative charge at physiological pH (as does cysteine, with a pKa of the sulfhydryl at 8.18). GSH oxidizes spontaneously at physiological pH to form the disulfide, oxidized GSH. Alternatively, GSH can react with cysteine or protein sulfhydryls to fbrm mixed disulfides. The role of GSH in the overall regulation of enzyme function is an area of much current study. "Essential" protein sulfhydryl groups are ubiquitous, whether in a catalytic role or in determining protein confbrmation, as is indicated by the inactivation of a large nunber of these enzymes by oxidants such as diamide, N—ethylmaleimide or 5,5'-dithiobis Figure 2: Chemical structure of glutathione CIOOH HzN-CIZ-H CH2 CH2 HN HS- cuz-C:H c=o ~+ CH2 c'oow GLUTATHIONE 1-L-glutamyl - L-cystoinylglycine (2-nitrobenzoic acid). For this reason, reducing agents, such as 2-mercaptoethanol or dithiothreitol are commonly used to stabilize enzymatic function. In light of the effects of thiol/disulfide exchange on enzyme activity, this covalent modification may play an important role in the biological control of various metabolic pathways. Further, the thiol/disulfide status of GSH may modulate enzyme activity [50], and recently, GSH disulfide has been shown to inactivate, destabilize, and enhance proteolytic V susceptibility of fructose-1,6-bis-phosphate aldolase [104]. The importance of GSH is underscored by its ubiquitous occurrence in every organism examined. GSH is a cofactor, coenzyme or substrate for an encyclopedic number of enzymatic reactions. On the molecular level, the most conspicuous aspect of cellular GSH depletion is its relationship to cell damage. That GSH provides an important cellular defense against oxidant injury has been well documented [16,68,133]. During a state of GSH deficiency, reactive metabolites, formed .12 ‘sitg, bind to macromolecules of biologic importance and cellular toxicity results [50]. The formation of oxygen-dependent free radicals and peroxides result in the peroxidation of membrane polyunsaturated fatty acids. 2. Enzymatic synthesis: The synthesis of GSH occurs in a series of 6 reactions of the Y-glutamyl cycle from precursor amino acids (Figure 3, from [92]). The enzymes of this cycle have been reviewed [94,96]. The availability of the precusor amino acid, cysteine, is the rate-limiting factor in GSH synthesis. The physiologic concentration of cysteine is about an order of magnitude lower than the Km of Y-glutamylcysteine synthetase for cysteine [114]. According to the Figure 3: Y-glutamyl cycle R-CH-COOH NH; Ammo and (ounce can ., II 74 M7; ”.7 I ’ ¢ ,, ///I/} Inn/.57 M, m’ .m. MHz/757 W/‘xxnzmozlfl/W/fl I’l” ,. /’I///II//, ”I I uCELl. usueuué I” w 5v: 5, ,; ‘1/7- -oLuuu\IL ’ ‘ mausrsenmse /// I/ // , I I57}; (1." l/' "' 7/' ','.'//"// 1.9.: /. $431141, ”fly/l ' -,.’ J 1557/0/17), /////////’([77///)// [Ml/m /.' "Iii/17 ’/// {4’1/1'7 III” 'I/ 2" .. . —- —- " " " ' m “7"" RECOGNHION ' _——" l - u- u- .. ”— "YRANSLOCMION" co " c co " og-eoon l1 ) a 3:":‘9‘ ‘f’fi’: y-glutamy‘ - "stony! -glycm 7’55-~H-é,.-coo,. N,N-CH-co-~u-CN,—c00n Ono-u”, [glutathione] I------_- ”9"?” ,. _--“ cystomyI-glyeme COOH “955315.". i"“f 0‘4"”: L". 3'3f'.".".":::‘° m ADP . r. LCOOH J I PEPYIOASE 1 """ [owwwnowe SVNMEYASE I z-GLUIAMVL CYCLOTPANSFERASE j /s m ”hem-coon ' "' R-(EH-COOH glycine 95-5" ”“2 co-uu-cu-coou .. "515‘“ N ' - " ‘ Amino and tar-Sn '09: [2) m.,," coll) Ngu-CH-COOH\ CH-NH )- gluilmyi ~cystome 7 Cyfllfifl' Ont-00,. m SCXOF‘WOL__I [NASE ‘0' . n S-o-eztomo “‘38: [7- owmavr- -Orerm£ SYNTHETA SE] éH'm' up ‘1' ADP o H swim and “RECOVEIY' (1| , NH 5! 10 report of Beatty and Read [l4], cysteine or cystine supported GSH synthesis in isolated rat hepatocytes better than methionine. However, the determination of GSH synthesis by these authors, which was by the rate of accumulation of cysteine-glutathione mixed disulfide in the incubation media, probably resulted in erroneous estimates of GSH synthesis, due to inhibition of GSH efflum from isolated rat hepatocytes by methionine [7]. Also, methionine, a precursor of cysteine via the cystathionine pathway, has been shown to be a good precursor fOr rapid GSH synthesis in hepatocytes [13,113]. The kidney differs from the liver, in that cysteine, as cystine, is readily taken up by tubular epithelium, supporting the requirements for the rapid turnover of glutathione in kidney [98,106], whereas methionine is poorly supportive of rapid GSH synthesis, probably because of a deficiency of cystathionase [98]. GSH regulates its own synthesis by non-allosteric competitive inhibition of the Y-glutamate binding site of Y—glutamylcysteine synthetase [39,114]. The y-glutamyl bond in the tripeptide renders GSH resistant to degradation by «a-peptidases, however, this bond is susceptible to hydrolytic and transpeptidation reactions leading to products recoverable by the cell. GSH synthesis can be inhibited by buthionine sulfoximine (Figure 4), a potent and specific inhibitor of Y-glutamylcysteine synthetase [54,55,56,58]. L-2-imidazolidone—4-carboxylate inhibits the enzyme S—oxoprolinase [58]. Potent, irreversible inhibition of Y-glutamyl transpeptidase activity can be attained with AThlZS [61]. The structure-activity relationships of substrate analogs on the inhibition of the Y-glutamyl cycle enzymes have been studied [57]. 11 Figure 4: Chemical structure of buthionine sulfoximine. C00- 4- I H3N-C'I-H «I»... I: 0=lSl-CH2-C': -H NH CH2 CH3 BUTHIONINE SULFOXIMINE 12 3. Glutathione utilization: Intracellular GSH is converted to oxidized GSH by selenium—containing glutathione peroxidase (GPx), which also catalyzes the reduction of H202 and other peroxides. GPx is specific for its hydrogen donor, GSH, and nonspecific for the hydroperoxide [95]. Therefore, GPx catalytically detoxicates a range of substrates from H202 to organic hydroperoxides. Although GPx shares the substrate H202 with catalase, it alone can react effectively' with organic hydroperoxides as well. Selenium—independent GPx activity describes the peroxidase activity of glutathione transferase [88]. In male weanling rats maintained on a selenium-deficient diet, GPX activity fell to undetectable levels. When these animals were also deprived of vitamin E and sulfur-containing amino acids, a fatal hepatic necrosis deve10ped [63,116]. The reduction of oxidized GSH, with concomitant oxidation of NADPH, is catalyzed by the flav0protein glutathione reductase (GRed) in an essentially irreversible reaction, accounting for the very high GSH:oxidized GSH ratios found in cells [95]. The importance of this enzyme is most apparent in cells which no longer synthesize macroomolecules, such as the mature erythrocyte where maintainance of adequate levels of reduced GSH is essential fOr the deformability and flexibility of these cells [71]. In rat hepatocytes, the protective role of GRed against adriamycin-mediated toxicity was demonstrated by the inactivation of this enzyme with BCNU [8]. In these experiments, the rapid recycling of oxidized GSH to GSH by GRed ameliorated the lipid peroxidation seen when GRed was inactivated. Glutathione S—transferase performs the catalytic function of conjugating 13 potentially harmful electrOphilic compounds of hydrophobic character ‘with the nucleOphilic GSH to produce a compound less toxic and more water soluble than the parent compound. By reason of their high affinity and concentration, they may also act as scavengers for alkylating agents. In a suicide—manner, the covalent bond fbrmation between the transferase and the highly reactive electrophile detoxicates the compound, at the expense of further enzyme function [73]. RATIONALE The overall research objective for studying biochemical differ- ences between the host and parasite is the possibility that this approach could lead to the rational development of new families of drugs which exhibit antiparasitic activity at levels of drug which are not toxic to the host. The rationale fbr investigating the mechanism of action of oltipraz is to determine the biochemical differences between these organisms which result in schistosomicidal activity with only minor side effects noted in the host. This may, in turn, lead to the developnent of better, safer and less expensive new chemotherapeutics . 14 MATERIALSANDMEIHODS A. Preparation of host and parasite tissue: Adult Schistosoma mansoni, 45-55 days post-infection were dissected from the hepatic and mesentaric veins of Swiss Webster female mice by the method of Fetterer _e_§ _a_l_. [45]. Where male parasites were used, worms were dissected into media containing 0.05% sodiun pentobarbital, mechanically separated and the males placed into fresh media and maintained at 37°C until assay. For _i_r_1_ vivo experiments, mice were dosed by gavage at either 125 mg/kg or 250 mg/kg oltipraz in peanut oil or peanut oil vehicle and sacrificed at various times post dosing. For anaerobic experiments, 40 male parasites were incubated 18 hours at 37 °C in 25 ml PIS/RPMI. Prior to incubation and inmediately after addition of worms to the media, nitrogen gas was passed through an oxygen trap (0.2 fl pyrogallol in 0.2 E NaOI-I) , then bubbled through the media to replace dissolved oxygen. For egg-laying experiments, fifteen pairs of animals were transferred at random to 50 m1 of media in 250 ml Erlenmeyer flasks. The medium was a 1:1 mixture of RPMI 1640 and heat inactivated horse seruu, to which had been added 100 U/ml penicillin and 100 ug/ml streptomycin (media components from Gibco Laboratories, Grand Island, NY). Mercaptoethanol was added to a final concentration of 50 pg. Drug or vehicle were added to the media in a volune not exceeding 100 15 16 ul. Flasks were then incubated for 72 h at 37°C, with continuous mechanical agitation. B. Glutathione assay: At the end of the incubation period, worms were filtered, weighed and homogenized (1:10, w/v) in 6% trichloroacetic acid (TCA). Host tissues were perfused ‘with ice—cold 1.15% KCl to clear blood from the organ and then homogenized 1:20 (wzv) in 6% TCA. Aliquots were removed fOr protein determination by the method of Albro [2]. Following 1000 x gcentrifugation, GSH was determined by a modification of the method of Ellman [44], where to 100 pl aliquots of supernatant were added 800 pl of 0.3 g NaZHPO4, pH 8.2 and 100 pl 0.04% l—fluoro—2,4 dinitrophenol. Absorbance was recorded at 412 nm for freshly prepared standards and samples. C. Manipulation of schistosome GSH levels in vitro: Schistosome GSH levels were altered by treatment of male parasites with diamide [88]. Tb determine the extent of lipid peroxidation, malondialdehyde, which is fbrmed as a breakdown product of polyunsaturated fatty acids was measured by the thiobarbituric acid assay of Buege and Aust [29]. Parasite GSH levels were measured after incubation in varying concentrations of BCNU (1,3-bis [Z-chloroethyl]-l-nitrosourea) for 3 h or 880 (L-buthionine—S-R-sulfOximine) fbr 6 h. The dose-dependency of oltipraz in depleting parasite GSH levels was established fbr l h, and the antagonism of oltipraz depression of schistosomal GSH by various compounds was evaluated at 18, 36 and 72 h. 17 D. High pressure liquid chromatography (HPLC): Male schistosomes were transferred to wells (multi—well tissue culture plates, Flow Laboratories, Inc., McLean, VA) containing 2.5 ml RPMI 1640, 3 x 105 dpm/well [3581cysteine or [3581cystine or [14C]glucose (Amersham Corp., Arlington Heights, IL) and other excgenously added components as indicated. Plates were maintained at 37 C in a dark chamber, for the time periods specified. Parasites were weighed and homogenized 1:10 (wzv) in 3.5% perchloric acid. The homogenate was centrifuged 12000 x g, 10 min. One-half ml aliquots of acid-soluble supernatant were derivatized and the derivatives analyzed by HPLC as described by Reed .g£.§l. [114]. Eluant was monitored at 352 nm and collected in 0.5 m1 fractions. Five ml ACS (Amersham Corp., Arlington Heights, IL) was added to each fraction and fractions were assayed for radioactivity by liquid scintillation (Beckman Instruments, Inc., Fullerton, CA). E. .Enzymg’ .assays: The enzyme activity of Y-glutamyl transpeptidase (Y-GTP) was determined according to a modification of the method of Orlowski and Meister [108]. Tissue homogenates (1:20, w:v) were prepared in 0.1 fl KZHPO4, 1 mg GSH, pH 8.0. A 20 pl aliquot of the supernatant fraction (25,000 x g, 20 min) was added to 480 pl substrate buffer consisting of 5.55 m! L-Y—glutamyl—p—nitroanilide, 11.1 m]! litJCl2 and 110 mg Tris-HCl. E‘nzymic activity was monitored at pH's from 6.0 to 9.5 in increments of 0.5 pH unit. The reaction was stopped with 1 ml 1.5 g acetic acid and the p—nitroaniline product formed was measured at 410 nm against a reference solution. Enzyme activity is expressed as uncles p-nitroaniline formed/min/ mg protein. 18 Glutathione S—transferase (GTr) activity was examined in tissue cytosol fractions, prepared as above, in the presence of 1m}! (SH, with 1,2-dichloro-4-nitrobenzene or p-nitrobenzyl chloride as substrate. The initial velocities of glutathione conjugate formation were measured spectrophotometrically at ambient temperature according to the procedure of Habig et 31. [64]. Specific activity is expressed as nmoles product/min/mg protein, after correction for spontaneous nonenzymatic conjugation. Glutathione reductase (GRed) activity was assayed by the method of Beutler [l7]. Either 50 pl of 2 mg NADPH (or NADH) was used as substrate and the rate of change in optical density at 340 nm was recorded. The oxidation of 1 mole NADPH (or NADH) per minute was used as a unit of GRed activity. Glutathione peroxidase (GPx) was measured by both direct and indirect ° assay methods. Both assays measure seleniun— and nonselenium—dependent GPx. The direct assay was performed by the method of Mills [100] as modified by Hafemangtal. [65]. Parasite tissues were prepared as described for the Y—GTP assay (rather than 4 volunes 0.15 _t_4_ KCl used by Hafeman _e_t_:_ El) . The indirect assay for the measurement of GP): was by the method of Beutler [18] . The rate of for- mation of oxidized GSH was measured by the change in absorbance upon the oxidation of NADPH to NADP+. 19 F. Mechanical, surface electrical and tegumental mtential measurements: Recordings of longitudinal muscle tension and muscle activity were made from male schistosomes as previously described [45] . Surface electrical activity was recorded by the use of suction electrodes as described by Semeyn gt _a__l_., [122]. Recordings of tegu- mental potentials were obtained by using glass microelectrodes as described by Thompson _e_t_: 511. [129]. CHAPTER ONE: HOST-PARASITE DIFFERENCES IN THE REGULATION OF GLUTATHIONE Introduction: Schistosome glutathione (GSH) levels declined after .in vivo treatment with the antischistosomal drug, oltipraz, while the GSH content of various host tissues was observed to increase [26], suggesting the possibility that there may be chemotherapeutically vulnerable sites in the parasite relating to biochemical differences in GSH metabolism. The relationship of GSH levels to the maintenance of schistosome viability has not been examined, although GSH has been detected in relatively high concentrations in almost all cells of living organisms, including the schistosomes. Presence of the enzymes of the Y—glutamyl cycle which regulate GSH synthesis have been reported only for the cestode Moniezia benedeni [90]. Because of its reactive sulfhydryl group, GSH is involved in a large variety of chemical reactions. The GSH S—transferase family of enzymes utilize GSH to protect proteins and membranes against reactive electrophilic species by the fOrmation of GSH adducts. GSH serves to protect cells against peroxides formed during oxidative metabolic processes in a GSH peroxidase catalyzed reaction [4]. Further, in addition to its role as cofactor fOr numerous reactions of cellular metabolism [48,81], GSH regulates the redox status of protein thiol groups, thus modulating various metabolic processes and membrane events [51,106], underscoring the importance of this tripeptide to both host and parasite. The enzymatic regulation of GSH by §;. mansoni was investigated in this report. 20 21 Results: Diamide effected a dose-dependent depletion of schistosome intracellular GSH (Figure 5). The production of'malondialdehyde, however, was not observed after 1 h incubations of parasites in diamide at concentrations as high as 3 my, even though the parasites were moribund at this concentration. Male §;_ mansoni possess low levels of the enzyme Y-glutamyl transpeptidase. The parasite enzyme activity (68 nmoles product formed/min/mg protein) is of the same order as uninfected mouse liver tissue (59 pmoles product fOrmed/min/mg protein). Uninfected mouse kidney cortex preparations exhibited greater activity, 331 umoles product fOrmed/min/mg protein. The pH optimum fOr parasite enzyme activity, pH 8.5, is the same as that observed fer the host tissues. Male parasites possess GSH S-transferase activity, with an optimum for catalytic activity at pH 8.0. The activity of the enzyme occurs with a.slight1y higher Km.than host liver, but the reduced affinity for substrate is not due to inhibitory factors in the parasite cytosol, as the cytosolic fraction did not inhibit GTr kinetics (purified GTr from equine liver, Sigma), although specific activity did decline over time, suggesting the presence of proteases in the parasite supernatant. The GTr activities of male and female parasites and of uninfected mouse liver are shown in Table 2. 22 Figure 5: Dose-dependent depletion of schistosome glutathione by diamide. Twenty adult male schistosomes were incubated in 2.5 ml RPMI 1640 containing various concentrations of diamide, as indicated. Incubations were performed at 37°C, in a dark chamber for 1 h. values represent two or three separate experiments with triplicate groups of 20 animals per group. vertical lines are‘: l S. E., Asterisk = p < 0.05. .x. u L .x. .. O R T. N 0 C 1.. 3i 2w 1.. O O O O O T- 2.39:. a: . $.5sz ~20. 5330 3.0 |.O .03 DIAMIDE (MM) 23 Table 2: Glutathione S-transferase activities of Schistosoma mansoni and uninfected mouse liver tissue. Substrate péNitrobenzyl Chloride l-Chloro—2,4-Dinitrobenzene Liver 199126 12101200 §;_mansoni male 26: 7 526:169 §;_mansoni female 9: 4 * 172:141 * Glutathione S—transferase activity is expressed as nmoles product/min/ lug protein, after correction fOr spontaneous nonenzymatic conjugation. Male _S_._ mansoni possess 10.3 i 0.2 n'U/mg protein GRed activity, which is 15.1% of mouse liver enzyme activity with NADPH as cofactor. ‘With NADH, GRed activity was reduced 90% in both parasite and mouse liver tissue. The presence of inhibitory factors in the schistosome homogenate was considered unlikely, as boiled worm homogenate did not inhibit the activity of purified yeast GRed (obtained from Sigma). Male schistosome GPx activity was not significantly above blank values with either the direct or indirect assay. Discussion: In this study, reduction of parasite GSH by diamide treatment .in .XiEEQ. to 30% of control did not yield a measurable increase in malondialdehyde equivalents. GSH depletion to a critical level, about 20% of control, was required before an increase in lipid peroxidation was observed in rat hepatocytes [136]. As the parasites were dead or dying at diamide concentrations effecting 60—70% GSH depletion, it would appear that diamide toxicity may not be dependent on membrane damage due to lipid fragmentation occurring as a result of peroxide attack. The toxicity observed fbr diamide may be attributed to nonspecific sulfhydryl oxidation [67]. Further, membrane proteins begin to fOrm intrachain disulfides in cells in which GSH has been depleted by 60-70% [89], resulting in altered membrane properties [112,124]. 25 The values obtained fOr Y-glutamyl transpeptidase activity from parasite supernatants may be misleading, since no ultrastructural studies on the localization of the enzyme were perfbrmed and the specific activity reported is fer soluble protein from the entire animal. In general, higher enzyme activity is located on the external surface of cells which exhibit prolific secretory or absorptive functions. Therefbre the enzyme may be localized to the tegument of the schistosome, which comprises only 1-2% of the total worm protein (unpublished results). As tissues are essentially impermeable to exogenous GSH without the transpeptidase [66], these results suggest that schistosomes may be capable of absorbing GSH from the host blood stream, possibly coupled with amino acid uptake by the classical Y-glutamyl cycle [97,99]. Schistosomes are capable of performing phase 2 conjugation reactions, suggesting that GTr activity'may play a role in detoxication of chemotherapeutic agents. Female parasite activity at 30-40% of male GTr values is without explanation, however, other sex-related differences in the activities of schistosomal enzymes have been observed [37]. Further, the large amount of soluble protein released from the female digestive system during homogenization. may' have resulted in lower specific activities than are actually present intracellularly. 26 That schistosomal GRed activity was only 15% as active as mammalian liver tissue could indicate that the parasite is more at risk to oxidative damage when GSH levels are depleted. However, in light of the fact that §;_ mansoni are functional anaerobes [27], the low level of GRed activity is not surprising. The level of GRed would still be the primary determinant of the availability of reduced glutathione because GRed is considered to be the rate limiting factor in the redox cycling of GSH [104,125]. The results with the two pyridine nucleotide substrates on host and schistosomal GRed activity suggest that the enzyme is NADPH specific and that there may be one or more transhydrogenases present in the crude homogenate capable of transferring the reducing equivalent from NADH to NADP+. The importance of functional mitochondria to the maintenance of control levels of reduced GSH in rat tissue was shown by Jocelyn [78], where although the reduction of GSH by diamide was mediated by GSH oxidation, GSH levels were maintained due to regeneration by NADH driven transhydrogenase and NADP+-specific GRed. The functional status of schistosome mitochondria has not been clearly elucidated. The results obtained in the present study showing no lipid peroxidation in the presence of diamide suggest that the activities of parasite GRed and transhydrogenase reactions are adequate to the task of rapidly recycling GSH from oxidized GSH in the face of an oxidant Stress o 27 That the lack of significant GPx activity in the male schistosome is compatible with parasite viability indicates that under physiologic conditions either the parasites are not exposed to peroxide stress or have evolved other mechanisms to minimize or repair peroxide-induced toxicity. Circulating concentrations of lipid peroxides in the blood stream are negligible, likely due to the high GPx activity reported fbr host red cells [17]. Endogenous peroxide formation is probably minimal in schistosomes as reports on parasite metabolism indicate a reliance on reductive or conjugative reactions fOr drug metabolism [41,42,43,l03]. Also, schistosomes do not possess a functional cytochrome mixed function oxidase system [10], further minimizing the level of toxic insult to the parasite due to GPx deficiency. Furthermore, there is a nonenzymic decomposition of peroxide by GSH, ‘proportional to GSH levels [100], which may provide adequate protection for the parasite from low levels of intracellularly generated peroxides. A. chemotherapeutic possibility, the inhibition of ‘v-glutamyl transpeptidase would prevent the normal extracellular breakdown of GSH and inhibit translocation of Y-glutamyl amino acids intracellularly. In mice treated with AT 125, an irreversible Y-glutamyl transpeptidase inhibitor, and in two human patients exhibiting congenital Y-GTP deficiency, glutathionemia and glutathionuria developed, with marked enhancement in the renal excretion of Y-glutamylcysteine, cysteine and cystine also being observed [60,63]. The toxicity of irreversible inhibitors of Y-GTP to the schistosome have yet to be studied. Therefore, while enzymatic differences between host and parasite were 28 observed in this report, the possibility for the rational design of a selective chemotherapeutic compound based on these differences is marg inal . CHAPTER TWO: ACUTE EFFECTS OF OUTIPRAZ AND ITS ANTAGONISMIINTVITRO Introduction: Bueding e_t_ 31. [26] observed a depression of parasite glutathione (GSH) content as one of the earliest biochemical changes after adminis- tration of oltipraz _i_n 2.13.9: As the reduction in parasite GSH was accompanied by an increase in host tissue levels of (BH, the possibil- ity for the rational design of a drug selectively toxic to the parasite based on interference with parasite glutathione metabolism or synthesis suggests itself. Tb further define the relationship between the antischistosomal activity of oltipraz and parasite (SH levels, the acute effects of oltipraz on various physiological parameters of S:- mansoni 3:9. vivo and _i_n_ vitro are presented. Results : _I_r_1__y_i_v_o_, oltipraz (250 ng/kg) , effected a significant reduction in male parasite, host liver and host renal cortex GSH levels by 1 h, with maximun depression at l h for host tissues and 3 h for parasite tissue (Figure 6). (EH levels of the host rebound to or above control levels at 6 h. However, parasite GSH levels remained significantly depressed at that time. in y_i_t_r_g, oltipraz (10 pg) significantly depressed schistosome GSH levels by l h, (Figure 7) an effect not observed when parasites were coincubated in GSH (1 my.) , cysteine (1 my) or methionine (1 It!) . Neither UI'I‘ nor 2-mercaptoethanol (100 p]! or 1 ml!) blocked the oltipraz-induced depression of schistosome GSH in vitro (Figure 8). 29 30 Figure 6: Oltipraz effect on glutathione content in infected mouse hepatic and renal cortical tissue and in male schistosomes _ig 23119. Infected mice were dosed with 250 mg/kg oltipraz in peanut oil by gavage. Mice were allowed food and water ad _l__i_b. Worms from 6 mice were pooled and GSH was assayed from groups of approx. 40 males [0]. Tissue slices of host liver [I] or kidney cortex slices [9] were pooled and 5 groups of approx. 100 mg (wet weight) tissue were assayed for GSH: Asterisk = p<0.05 from time 0 levels. A7520... 9.: . 3.0:. 5 .30 Hours 31 Figure 7: Dose-dependent depletion of schistosomal glutathione by oltipraz. Twenty adult male schistosomes were incubated in 2.5 ml RPMI containing various concentrations of oltipraz, as indicated. Incubations were performed at 37°C, in a dark chamber, for 1 h. Values represent two or three separate experiments with triplicate groups of 20 animals per group. Vertical lines are i l S.E., Asterisk = p <0.05. *lO' C ONTROL \ *IU' 30 q I a O. W O O b-2585 a: 3825 50.1.2540 OLTIPRAZ (pM) 32 Figure 8: Effect of oltipraz and various compounds on schistosome glutathione levels in 17.3.29: Paired schistosomes were incubated in 50 m1 HS/RPMI at 37 °C, in a dark chamber with continuous mechanical agita- tion. [as] = p < 0.05 from control value at the same time point (without oltipraz control). [*1 = p < 0.05 from oltipraz value at the same time point. + oltipraz is schistosomes incubated in 1 [1M oltipraz. [c] is glutathione, reduced form; [I] is cysteine (1 m_M_); [c] is methionine (1 mg); [fir] is dithiothreitol (100 u!) and [*1 is oltipraz (1 u!) . Values represent means of triplicate samples of males which were separated from female parasites at the end of the experi- ment . GSH (nmoles-mg protein" ) .40‘ as 9 to 9 With Oltipraz Without Oltipraz O 0 3 z} ‘4 I l I _"—" 18 36 72 72 TIME (Hrs) 33 Surprisingly, oltipraz (l u!) lowered parasite GSH levels only in an aerobic environment. 10 u! oltipraz produced a more pronounced effect, but again, only under aerobic conditions (Figure 9). 35S]cysteine or [3SS]cystine by male By one h, the uptake of [ schistosomes was significantly inhibited (41% and 36% of control, respectively) during 12 vitro incubations in the presence of 5 1g! 35S]GSH formation occurred in the presence of 5 oltipraz (Figure 10). [ uM oltipraz, and although less label was incorporated, the relative distribution of either labelled precursor was minimally affected. There were no significant differences between the cysteine and cystine controls. [14C1glucose uptake was significantly reduced by 5 u! oltipraz only at 60 min (Figure 11). Neither the oxy derivative of oltipraz (100 pg) nor cysteine (1 m!) had any effect on labelled glucose uptake. The amplitude and frequency of endogenous electrial transients in male §;_ ‘mansoni were profoundly depressed after in yiyg exposure to oltipraz, even at the low dose of 125 mg/kg. The onset of significant depression occurred by 6 h, with the high amplitude (>40 uV) potentials exhibiting a higher level of sensitivity (Figure 12). Schistosomes exposed to 3.5 uM_ oltipraz ‘in .XiEEQ. exhibited a similar level of depression to the drug, with high amplitude electrical activity being significantly depressed after incubations as brief as l h. The profbund depression of surface electrical activity observed after 24 h incubations in 10 HM oltipraz was not observed when the parasites were 34 Figure 9: Oltipraz effect on schistosomal glutathione levels in yitgg: Aerobic vs anaerobic environment. Paired schistosomes were incubated as described in Figure 8 in either an aerobic or anaerobic environment, with or without oltipraz, as indicated. Fbr anaerobic experiments, N2 first bubbled through an oxygen trap occupied the gas phase in the flasks. values represent the means of triplicate samples of males separated at the end of the experiment. [at] = p < 0.05 from control value at the time period specified, [as] = p < 0.05 anaerobic value significantly different from corresponding aerobic value. vertical lines are l S.E. GSH (n moles/mg protein) 29 so Air Control "2 18" ‘°’°M WW 4;:— lO-sM Oltipraz 1:; * * 36H * Card 112‘- E 10‘6M Oltipraz 1:; 3 Figure 10: [ 5S]Cysteine uptake and incorporation into schistosomes in vitro. Male schistosomes were incubated in 2.5 ml RPMI 1640 containing 3x105 dpm [3SS]cysteine per well at 37°C, in the dark, in the pres- ence of 5 u! oltipraz (solid symbols) or vehicle only (empty symbols). [ c or o ] represent total uptake (disintegrations/min/male) of [BSSlcysteine, solid line; [:3 or I ] represent the relative percentage of total 358 as [3SS]GSH and oxidized [3SS]GSH, broken line. 10 males per well, values represent means of 4 wells per data point; Asterisk = statistically different (p<0.05) from control uptake. Relative distribution of label, once assimilated by the parasite, into [3SS]GSH and oxidized [3SS]GSH was not statistically different from control. I I I .I I I I I 'I I I I d O o 5 4 30 400 20 (hoard TIME 36 Figure 11: [14C1Glucose uptake into schistosomes in 111559. Male schistosomes were incubated as‘ in Figure 10, except wells contained 3 x 105 dpn [14C]glucose per well. [0] represents untreated males, [I] represents oltipraz-treated animals (5 11M) , [n] represents parasites treated with the oxy analog of oltipraz (50 11M) and [0] represents male schistosomes coincubated in 5 pp} oltipraz and 1 m_M_ cysteine. Values represent means of 4 wells per data point; Asterisk = p<0.05. dpm ~ molo "' 60 8 8 N 0 IO 3'0 TIME (min) I 40 I 50 60 37 Figure 12: Surface electrical activity of male _S_._ mansoni __i_n X119: Oltipraz effect. Mice (55 days p.i.) were dosed by gavage with 125 mg/kg oltipraz [I], 250 mg/kg oltipraz [I] or peanut oil vehicle [0] and sacrificed at the times indicated. Values presented are the means of 6 to 8 animals analyzed per time point per test dose; vertical lines are l S.E. Asterisk = p<0.05. 7.2.33 . 32:3.oo TIME (Hrs) 38 coincubated in 1 my cysteine or 1 mg glutathione, but not by l m}! or 100 RM 2-mercaptoethanol. The oxy analog of oltipraz (100 u!) did not alter parasite surface electrical activity from control levels. Tegunent potentials recorded from male schistosomes were significantly depolarized by l u! oltipraz following 18 h in yitrg incubation. This effect was prevented by the addition of 1 ml} cysteine to the incubation median, whereas cysteine by itself did not alter the tegument potential, nor did the oxy analog of oltipraz (Figure 13) . Discuss ion: Bueding st 31. [26] observed that _i_n 11332 treatment of S_._ mansoni with a 250 mg/kg dose of oltipraz resulted in a depression of parasite GSH levels to roughly 40% of control over the first week after treatment. In the present study, depletion of GSH was observed by l h in both parasite and host tissues at a dosage of 250 mg/kg peg 2s. The fact that GSH levels in parasite tissue were not rapidly restored to control levels, as was seen in host tissues, indicates that there may be important differences in the biochemical regulation of GSH between the parasite and host. The present results are not in conflict with those of Bueding _e_t_ _a_Il_ which showed depressed parasite GSH and elevated host tissue GSH after oltipraz treatment _i_n lilo, as their experiments did not include measurement of the acute response of host tissue or parasite GSH levels to oltipraz. A similar depletion of parasite GSH levels in the presence of oltipraz was observed __i_n_ Egg, allowing the acute actions of oltipraz on the parasite to be studied independent of 39 Figure 13: Tegunent potential of male schistosomes .1-2 1122?: Effect of Oltipraz and antagonism of effect by cysteine. Male schistosomes were incubated in HS/RPMI containing Oltipraz or oltipraz plus cysteine as indicated, or in the presence of 100 pl! oxy-oltipraz, for 12 h at 37 °C, in the dark. Values presented are the means of 5 animals per tegu— mental potential average, vertical lines are 1 S.E. -L 1 Control * SID-GM Oltipraz * -{::]10‘5M Oltipraz -L 110-506 Oltipraz + IO'aM Cysteine L IIO'aM Cystoino -[ JIO-4M Oxy-Oltiproz l l -60 -55 -so -45 -4o Togurnont Potontiol (mV) 40 the variables of host reaction to the drug. Bueding _e_t_ _a_1_. speculated that the chemotherapeutic activity of oltipraz may be due to competition of the drug with Y-glutamyl cysteine, a precursor of GSH synthesis. By this mechanism the non-stoichiometric depletion of GSH could occur at a rate dependent on the degree of inhibition of synthesis and the turnover rate fOr parasitic intracellular GSH. The results of the present study suggest that there may be an alternative explanation fOr the effect of oltipraz on GSH levels. First, the inhibition of cysteine uptake by'5 p! oltipraz corroborates results by Seed (personal communication) that uptake of both cysteine and glutathione, but not other amino acids, is retarded by oltipraz. Secondly, male schistosomes were able to incorporate [3581cysteine label into the glutathione pool, as GSH and oxidized GSH, in the presence of 10 pg oltipraz, at a relative percent not significantly different from control. Therefore, under the condi- tions of acute exposure, it is unlikely that GSH synthesis is being directly inhibited by oltipraz. That the oxy derivative of oltipraz, which has negligible antischistosomal effect, did not affect the uptake of [3SS]cysteine into the parasites, even at 50 pg, suggests that the thione group of the drug molecule is a necessary participant in the oltipraz-induced effects on transport mechanisms. 41 In the present study, GSH (l I!!!) , cysteine (1 It!) and methionine (1 my.) , but not 1 I!!! or 100 p! DI'I' or 2-mercaptoethanol, were able to block the _i_n 3.1.13.9. effect of oltipraz on GSH levels occurring during incubation. The antagonism of oltipraz effects by GSH or (SH precur- sors may be mediated through a process of mass action on the replenish- ment of GSH stores. DI'I‘ and 2-mercaptoethanol, unlike the other compounds tested, are incapable of participating in the production of GSH, which may explain their inability to antagonize the effects of oltipraz. Alternatively, the inactivity of the oxy derivative of oltipraz and the rather stringent structural requirements for the maintenance of antischistosomal activity of a series of oltipraz congeners [26] suggest that oltipraz may influence a membrane receptor, possibly functioning as a dipeptidase or transport carrier molecule for amino acids. The observation that oltipraz lowered parasite GSH levels only when the parasites were incubated in an aerobic environment indicates that it is probably an oxidative reaction that the (SH is participating in. Figure 14 illustrates a hypothetical scheme which could explain the oxygen-dependency of oltipraz effect on schistosome GSH levels. If oltipraz cycles a free radical, then organic components of the membrane could react with oltipraz to form an organic radical. In an oxygen-dependent reaction, an oxide radical is created which donates the radical back to oltipraz with the abstraction of a hydrogen, forming an organic peroxide. While schistosomes possess negligible GPx activity, GSH detoxication of lipid peroxides occurs nonenzymatically [100]. Similar radical cycling has been suggested for nitrofurans 42 Figure 14: Hypothetical reaction sequence to explain oxygen-dependency of oltipraz-induced depletion of schistosomal GSH ‘32, vitro. R represents an organic (presumably a lipid) component of the membrane, 0LT refers to oltipraz. GSH and G586 represents glutathione reduced and oxidized , respectively. NADH n- I 902‘ I transhydrogonuo RH GSSG NADPH HYPOTHETICAL REACTION SEQUENCE RELATING OLTIPRAZ TO GLUTATHIONE OXIDATION 43 [19], adriamycin and daunomycin [52] and paraquat [80]. Oltipraz-induced decrease in glucose uptake may be an indirect consequence of the drug's inhibitory effect on parasite motor activity. This is supported by the Observation that high amplitude electrical potentials, which appear to represent summed electrical activity originating from schistosome muscle tissue [122] , are reduced in frequency by the drug. Thiol interference by Oltipraz could also result in the inhibition of glucose utilization because of the wide range of enzymes that require a thiol group as cofactor for their cata- lytic activities [52] . The fact that tegumental morphology is not affected by _i_n y_i_t_r_o treatment of the parasites with 10 p_M_ Oltipraz for 12 h [23] indicates that structural damage to the tegument is not a contributory factor in the induction of acute oltipraz effects on membrane transport. mile the structural integrity of the outer tegumental membrane was maintained, microelectrode recordings showing depolarization of the tegument suggest that a significant redistribution of ions across the tegumental membrane had occurred. The effect(s) of a drug-induced ionic imbalance on the Na—driven carrier mediated cotransport of glucose [131] may have contributed to the decrease in glucose uptake observed. In a similar manner, perturbations of ionic equilibria across the tegument may contribute to the inhibition by oltipraz of the uptake of GSH precursors that was observed. Consistent with the effects on other parameters measured, tegument potentials of worms incubated in 1 11!} cysteine or 10 p! oltipraz plus 1 my. cysteine were 44 not significantly different from control values. These results indicate that cysteine is able to prevent Oltipraz-induced ionic imbal- ances .ig"yit£g_ without directly affecting ionic fluxes across the membrane. The fact that the oxy derivative of oltipraz did not affect the tegumental membrane potential further suggests that a disruption of membrane conductance may contribute to the antischistosomal efficacy of Oltipraz. Oltipraz is a very lipophilic compound which is rapidly accumulated by the membranes Of the parasite, suggesting that there is a hydrophobic nature to the site of drug action. Intercalation Of drug into the tegument and muscle membranes may' partially explain the effects Of Oltipraz on tegument potential and surface electrical activity, in a manner reflecting the correlation between the solubility of drugs in membranes and their ability to fluidize and disorder ‘membrane structure [110,121]. From a therapeutic perspective, the report by Ali .33 .gl. [3] which showed that coadministration of cysteine and Oltipraz to monkeys resulted in an enhanced bioavailability of the drug, suggests that the ‘ig .yiyg_ synergistic action of cysteine on oltipraz schistosomicidal effects may be due to this enhancement of oltipraz bioavailability. As the synergistic interaction of cysteine with Oltipraz may be indirect and removed from the site of drug action, the .in .yiE£g_ evidence presented which show cysteine antagonistic to Oltipraz action does not refute the $2 2129 Observations. Instead, these findings suggest that although cysteine, GSH or methionine ameliorate oltipraz action acutely 45 when applied directly to the parasite, in the long-term _i_n vivo situation, the enhancement of oltipraz bioavailability is more relevant clinically in the treatment of the disease. cm THREE: RELATIONSHIP OF SCHISTUSOME GLUTATHIONE LEVELS .10 VARIOUS PHYSIOLOGIC PARAMETERS OF THE PARASITE Introduction; Schistosome GSH levels were manipulated by various compounds in order to determine dose-dependency of acute effect and to determine if these effects were also antagonized by cysteine, methionine and/or glutathione in like fashion to the antagonism of oltipraz-induced GSH depression in the parasites. Results: BCNU, an alkylating agent which inactivates GRed [49], effected a dose-dependent depletion Of parasite GSH levels after 3 h incubation (Figure 15). Microelectrode recordings of schistosome tegu- mental potential revealed that ionic conductance is also altered by BCNU in a dose-dependent manner, an effect which is partially reversed by coincubation of the parasites in l m! cysteine (Figure 16). Tegumental potentials fOr 1 mM_ cysteine—treated control, fOr l m! cysteine plus 3 m! BCNU-treated parasites, and fOr 3 mg BCNU-treated animals were statistically different from each Of the other groups (P (0.91). 46 47 Figure 15: Dose-dependent depletion of schistosomal glutathione by BCNU. Wenty adult male schistosomes were incubated in 2.5 ml RPMI containing various concentrations of BCNU, as indicated. Incubations were performed at 37 °C, in a dark chamber, for 3 h. Values represent two or three separate experiments with triplicate groups of 20 animals per group. Vertical lines are i l S.E., Asterisk = p < 0.05. CONTROL A.-2_m.pozo oz. 330sz mzoib‘gu no no 0 3!) L0 133 (MM) BCNU 48 Figure 16: Tegument potential of male schistosomes _i_n_ 13319: Effect of BCNU and antagonism of effect by cysteine. Male schistosomes were incubated in HS/RPMI containing BCNU or BCNU plus cysteine as indicated, for 12 h at 37°C, in the dark. Values presented are the means of 5 animals per tegumental potential average, vertical lines are l S.E. -L 1 Control : lmM Cysteine + 3mM BCNU -l 3mM BCNU £22.3me BCNU L l L j l -50 '40 -3O -2O " IO Tegurnent Potential (HIV) 49 BSO, an inhibitor of Y-glutamyl cysteine synthetase [60], produced a dose-dependent depletion of GSH in male §;_ mansoni. The decline in GSH levels (Figure 17) was less dramatic than results obtained with diamide or BCNU and required a longer incubation period. NO acute differences from control were observed in longitudinal muscle tension development over the first 30 min of exposure to 1 m}! B80. B80 at 100 pH for 18 h produced a significant depression of high ampliture (> 40 pV) electrical potentials recorded from the surface Of male parasites (93 :_ 47 potentials ESQ—treated, 253 :_ 81 potentials control). Coincubation in the presence of l mM_cysteine inhibited BSD—induced depression of electrical activity (262 :.92 potentials). Surprisingly, Bsobtreatment at 1 mg resulted in egg counts not different from controls over the 72 h period, although GSH levels fOr the paired worms were depressed 30-40% from control levels at 72 h. Oltipraz effects on schistosome fecundity in yitgg are shown in Figure 18. The antagonism of Oltipraz effects by various compounds in the same assay is shown in Figure 19. Discussion: BCNU irreversibly inactivates GRed. This inhibition is selective as BCNU does not inhibit either the enzymes of glucose catabolism or GPx activity [49]. The prevention by cysteine of GRed inactivation by BCNU [49] may explain the antagonism of BCNU-induced depression of schistosomal tegumental potentials observed in this study, alternatively, as cysteine feeds into GSH synthesis, these data are also supportive Of the hypothetical reaction sequence shown in. Figure 50 Figure 17: Dose-dependent depletion of schistosomal glutathione by BSO. Twenty adult male schistosomes were incubated in 2.5 ml RPMI con- taining various concentrations of B80, as indicated. Incubations were performed at 37 °C, in a dark chamber, for 6 h. Values represent two or three separate experiments with triplicate groups of 20 animals per group. Vertical lines are i l S.E., Asterisk = p < 0.05. CONTROL * I I 3.0 [.0 I” o. O O 0.3 2. O A7339: 92.330sz $.25...st BSO (MM) 51 Figure 18: Oltipraz inhibition Of schistosome fecundity .ig_'yitgg. Paired schistosomes (15 pair per 50 ml HS/RPMI in 250 ml Erlenmeyer flasks) were incubated 72 h at 37‘C in the presence or absence of Oltipraz at the concentrations indicated. Asterisk = p < 0.05 from control. values presented are the means of 5 to 7 flasks run at each concentration; vertical lines are l S.E. No. of eggs per female in 72 hours o 59 I90 I§O 290 Control I'- Io‘7 M OI: 4xIO'7 M on as Io‘6M OI: * Figure 19: Effect of various reagents on oltipraz inhibition Of schistosome fecundity iguyitgo. Paired schistosomes were incubated as described in Figure 18. +OLT indicates coincubation of parasites in 0.4 pg oltipraz, McEtOH is 2— mercaptoethanol, DTT is dithiothreitol (Cleland's reagent), CYST is cysteine, METH is methionine and GSH is glutathione, reduced form. [*1 = p < 0.05 from control, [at] = P < 0.05 from Oltipraz only, i.e. statistically significant prevention of oltipraz effect. values presented are the means of 5 to 7 flasks run at each concentration; vertical lines are l S.E. No. of eggs per female in 72 hours L 5.0 19° "5.0 Control OI t * II 10% Menomon * 101M DTT * D" + O" * IO‘3M METH+Olt lO‘3M osn asu.on_——E‘” 14. Despite the toxic effects of BCNU on the schistosome, there is little chemotherapeutic potential for this particular compound. BCNU is an anticancer agent which exhibits mammalian toxicity due to alkyla— tion of biological macromolecules. Thus, while BCNU may selectively jeopardize the schistosome because of its absence of GPx activity, the carcinogenic potential of this drug would lhmit its usefulness. As BCNU appears to be able to inactivate GRed without metabolic activa— tion, its mechanism of action may not be related to alkylation, hence there may be potential fOr a BCNU-like compound if the enzymic inhibi- tion properties can be dissociated from the alkylation potential inherent in BCNU. BSO requires intracellular activation to BSO—phosphate before inhibition of Y—glutamylcysteine synthetase occurs. If the plateauing of GSH depletion at 6 h due to BSO concentrations greater that 1 mg indicates near total inhibition of the enzyme, the slow rate of GSH decline suggests that the turnover rate 12 21259 is on the order Of 12.5 h. TUrnover rates in the literature vary from estimations Of 0.5 h fOr rat kidney to 65-96 h for mammalian erythrocytes. Tissue culture and tumor cells exhibit biological half—life fOr GSH in the range of 6-8 h [87]. 54 Methionine, cysteine and GSH were observed to provide significant ameliorative action on egg laying inhibition due to Oltipraz. Both methionine and cystein e may be used fOr the intracellular enzymatic synthesis of GSH, which may aid in the maintenance of GSH levels, whereas DTT and 2—mercaptoethanol do not contribute to the synthesis of GSH. The poor correlation between GSH levels and fecundity was unexpected, however it may be an artifact of the technique employed, as worms from several flasks were pooled in order to assay for GSH. Alternatively, lg ZiEEQ culture dramatically affects oviposition in effecting a lower output of eggs, especially during the first 24 h period, when the worm pairs are "in shock" while they adjust to the new media and nonphysiologic conditions [unpublished observations]. As many factors may be affecting the reproductive system which do not influence GSH levels, the amelioration of oltipraz effects on schisto- some reproduction may be merely a qualitative finding. CHAPTER FOUR: EFFECTS OF SCHISTOSOMAL INFECTION ON HOST REGULATION OF GLUTATHIONE LEVELS Introduction: The cause of the major disease syndromes of schistosomiasis is the retention of a large number of eggs in host tissue [134]. Most of the eggs freed into the circulation are sieved out by the liver where host granulomatous inflammatory response to the eggs results in a presinusoidal blockage of portal blood flow, as well as hepatic necrosis and subsequent fibrosis. Monoamine oxidase activity, a sensitive indicator of liver fibrosis [72], is significantly depressed in mice by the 6th week following infection with §;. mansoni [1]. There is also a marked depression of microsomal drug-metabolizing enzyme activities of the host liver. A close correlation exists between the onset and degree Of enzymic depression with the severity and age of infection [32,33]. In the metabolism of xenobiotics, the tripeptide glutathione (GSH) plays an important role in chemical detoxication processes [52]. Experhmental GSH depletion exacerbates toxicity due to protein alkyla- tion or lipid peroxidation caused by chemically reactive, electrophilic chemicals or metabolites [4,77]. Glutathione peroxidase (GPx) is an integral part of the cellular antioxidative system [36], reducing peroxides in a reaction with GSH, which becomes oxidized in the process. Glutathione reductase (GRed) reverts oxidized GSH to its reduced state at the expense of NADPH. The glutathione S—transferase 55 56 (GTr) family of enzymes removes potentially cytotoxic alkylating agents through conjugation with GSH [21,84]. GTr also have GPx activity [113] which suggests a secondary role in detoxication of the byaproducts of oxygen utilization. As the toxicity Of antiparasitic drugs to the host may limit the therapeutic value of these compounds, changes in the ability of the host to detoxify xenobiotics due to the presence of parasitic infec- tions are of considerable importance. Results: GSH levels in host liver decline during the course of the infec- tion, while kidney cortex levels remain constant (Figure 20). Hepatic GSH is statistically depressed from uninfected liver values by day 50. Hepatic enzyme activities are summarized in Table 3. The GRed activity of infected mouse liver was not significantly different from uninfected controls at 50-60 days, however GPx enzymic activity was depressed to 37% Of uninfected levels. Hepatic GTr from infected mice exhibited 26% of uninfected liver activity. Neither renal GPx nor GRed enzymic activities were altered due to schistosomal infection. Kidney cortex was not examined fOr GTr activity. 57 Figure 20: Glutathione levels in host hepatic and renal cortical tissues during the course of Schistosoma mansoni infection. Means of triplicate samples of organs where the host harbored 15-30 pairs of parasites, n=5 animals per date, [I] represents hepatic GSH, [I] represents renal cortical GSH. Asterisk = statistically different (p<0.05) from uninfected control values at the time period specified, vertical lines are l S.E. nmoles GSH - mg protein ‘4 150 « 1.00 ~ 0.50 « 30 40 so 60 7o 80 90 AGE (Days Post—Infection) 58 Table 3: Mouse hepatic glutathione—related enzyme activity: Effect of Schistosoma mansoni infection. Hepatic enzyme activity GPx 1 GRed 2 GTr 3 Uninfected liver 440143 6617 199126 Infected liver 161139 * 62111 ns 52122 * 1 - Glutathione peroxidase activity, with t—butyl hydroperoxide as substrate, is expressed as nmoles NADPH oxidized/min/mg protein. 2 - A unit of glutathione reductase activity represents the oxidation of 1 pmole NADPH/min/mg protein, results are expressed as mU. 3 - Glutathione S—transferase activity is expressed as nmoles product per min/mg protein, after correction for spontaneous nonenzymatic conjugation, p—nitrobenzyl chloride was the substrate. * - Statistically different (p<0.05) from uninfected liver value, ns= not significant as determined by Student's t—test. 59 Discussion: The relative contribution of GSH depletion or the depression of hepatic enzyme activity to the hepatopathology of schistosomiasis could depend, in part, on the redundancy of the glutathione system. A func- tional reserve capacity fOr GPx [127] and GRed [49] has been shown in mice. Further, as the GTr are a family of enzymes with overlapping substrate specificities, and constitute 5-10% of the liver cytosol protein [25], a functional reserve capacity for mOuse liver GTr is also likely. In addition, y—glutamylcysteine synthetase activity' may increase as GSH levels decline [76]. However, the rate of synthesis of GSH in the infected animals was inadequate to maintain GSH at control levels, despite possible redundancy in the system. These results suggest that animals already compromised by a schistosomal infection may have enhanced susceptibility for hepatic toxicity from xenobiotic compounds which impose either an oxidative stress or require conjuga- tion fOr detoxication. SUMMARY AND CONCLUSIONS The present study indicates that the prolonged antischistosomal activity of oltipraz may be due, in part, to the acute effects observed .12 vivo and .15 vitro. Disruption of tegumental function in the transport of glutathione or glutathione precursors into the schistosome effects a depletion of intracellular GSH stores. As was observed fOr other sulfhydryl inhibitors [73], the ionic gradient across the tegu- mental membrane is disrupted by oltipraz. These effects of the drug on the tegument and the subsequent depletion Of schistosomal GSH lead to the depression of basic physiological parameters (frequency and amplitude of surface electrical potentials and inhibition of reproductive processes) and the alteration of biochemical processes, including indirect effects on glucose utilization. The present study also indicates that GSH and GSH precursors inhibit the acute actions of oltipraz '12 ‘yiggg. A likely hypothesis fOr the synergistic action of cysteine on the schistosomicidal activity of Oltipraz .12 .2129 is related to cysteine's enhancement of the bioavailability of the drug [3]. That DTT and 2-mercaptoethanol do not antagonize GSH effects .13 .yipgg is probably related, in part, to the fact that these compounds cannot serve as precursors to d9 novo GSH synthesis. 60 61 Concerning the enzymes of glutathione regulation, one of the major findings of this study is the elucidation of biochemical differences between the host and the parasite in the level of enzymic activity present in GPx, GRed, our and Y-GTP. These differences may be exploit- able chemotherapeutically. The fact that the parasite has different enzymic activities in regard to GSH utilization is not surprising because of the difference in the parasites metabolic handling Of xenobiotics compared to the host. Further, the parasites do not require oxidative reactions to provide energy, and therefOre, do not apparently require similar levels of GSH in order to protect against oxidant injury. In light of the hepato—pathology of the disease, changes in the biochemical regulation of GSH stores were not unexpected. These changes may indicate that the host is more at risk as the liver is the major organ for detoxication of most xenobiotics. REFERENCES 1. Abdel Samad, M. M., Guirgis, F. R., Zeitoun, R., El-Feki, R. F. and Awadalla, H. N. 1977. Liver monoamine oxidase (MAO) in liver homogenate of mice infected with Schistosoma mansoni and effect of certain therapeutic agents. Tropenmed. Parasit. 28: 554-559. 2. Albro, P. ‘W. 1975. Determination of protein in preparations of microsomes. Anal. Biochem. 64: 485-493. 3. Ali, H. M., Sulaiman, S. M., Bennett, J. L and Homeida, M. M. A. 1984. Effect of cysteine on Oltipraz blood levels in green monkeys (Cercopithecus aethiops). Chemotherapy 30: 255-261. 4. Anundi, I., Hogberg, J. and Stead, A. H. 1979. Glutathione depletion in isolated hepatocytes: Its relation to lipid peroxidation and cell damage. Acta pharmacol. et toxicol. 45: 45-51. 5. Asch, H. L. and Read, C. P. 1975. Transtegumental absorption of amino acids by male Schistosoma mansoni. J. Parasitol. 61: 378-379. 6. Asch, H. L. and Read, C. P. 1975b. Membrane transport in Schistosoma mansoni. Transport of amino acids by adult males. Exp. 7. Aw, T. Y., Ookhtens, M. and Kaplowitz, N. 1984. Inhibition of glutathione efflux from isolated rat hepatocytes by methionine. J. Biol. Chem. 259: 9355-9358. 8. Babson, J. R., Abell, N. S. and Reed, D. J. 1981. Protective role of the glutathione redox cycle against adriamycin-mediated toxicity in isolated hepatocytes. Biochem. Pharmac. 30: 2299-2304. 9. Barreau, M. Cotrel, C. and Jeanmart, C. 1977. German Patent 2,627,211. Chem. Abs. 86: 121,373h. l0. Barrett, J. 1981. Biochemistry .95. Parasitic Helminths. University Park Press. Baltimore. ll. Bartoli, G. M. and Sies, H. 1978. Reduced and oxidized glutathione efflux from liver. FEBS letters 86: 89-91. 12. Bauther, B. and Moyne, J. 1975. German Patent 2,430,802. Chem. Abs. 82: 156,258u. 13. Beatty, P. ‘W. and Reed, D. J. 1980. Involvement of the cystathionine pathway in the biosynthesis of glutathione by isolated rat hepatocytes. Arch Biochem. and Biophys. 204: 80-87. 62 63 14. Beatty, P. and Reed, D. J. 1981. Influence of cysteine upon the glutathione status of isolated rat hepatocytes. Biochem. Pharmacol. 30: 1227-1230. 15. Bennett, J., E. Bueding, A. R. Timms and R. G. Ehgstrom. 1969. Occurrence and levels of 5-hydroxytryptamine in Schistosoma mansoni. Mol. Pharmacol. 5: 542-545. 16. Bennett, J. L. and Depenbusch, J. W. 1984. The chemotherapy of schistosomiasis in Parasitic Diseases, Vol II. The Chemotherapy (Mansfield, ed.) pp 73-131. l7. Beutler, E. 1975. Glutathione reductase. In Red Cell Metabolism: A manual 93‘; Biochemical Methods 2nd ed. Grune and Stratton, New York, San Franscisco and London. pp. 69-71. 18. Beutler, E. 1975. Glutathione peroxidase. In Rep Cell Metabolism: _A_ manual pf Biochemical Methods 2nd ed. Game—5'53" Stratton, New York, San Franscisco and London. pp. 71-73. l9. Biaglow, J. E. 1981. Cellular electron transfer and radical mechanisms for drug metabolism. Radiation Res. 86: 212-242. 20. Bieder, A., Decouvelare, B., Gaillard, C., Depaire, H., Heusse, D., Dedoux, C., Lemar, M., Le Roy, J. P., Raynaud, L., Snozzi, C. and Gregoire, J. 1983. Comparison of the metabolism of oltipraz in the mouse, rat and monkey and in man: Distribution of the metabolites in each species. Arzneim.-Forsch./ Drug Res. 33: 1289-1297. 21. Boyland, E. and Chasseaud, L. F. 1969. The role Of glutathione and glutathione S-transferases in mercapturic acid biosynthesis . Adv . Enzymol . 32 : 172-219 . 22. Boyland, E. and Chasseaud, L. F. 1970. The effect of some carbonyl compounds on rat liver glutathione levels. Biochem. Pharmacol. 19: 1526-1528. 23. Bricker, C. S.,Depenbusch, J. W., Bennett, J. L. and Thompson, D. P. 1983. The relationship between tegumental disruption and muscle contration in Schistosoma mansoni exposed to various compounds. Z. Parasitenkd. 69: 61-71. 24. Bueding, E., unpublished studies 25. Bueding, E. and Bruce, J., unpublished studies 26. Bueding, E., Dolan, P. and Leroy, J. P. 1982. The antischistosomal activity of oltipraz. Res. Comm. Chem. Pathol. Pharmacol. 37: 293-303. 64 27. Bueding, E. and Fisher, J. 1982. Metabolic requirements of schistosomes. J. Parasitol. 68: 208-212. 28. Bueding, E., Naquira, C., Bouwman, S. and Rose, G. 1971. The antischistosomal activity of a nitrovinylfuran derivative (SQ 18,506) in mice and hamsters. J. Pharmacol. Exp. Ther. 178: 402-408. 29. Buege, J. A. and Aust, S. D. 1978. Microsomal lipid peroxidation. In Methods in Enzymology VOl. 52. Biomembranes Part C: Biological oxidations: MicrosomaI, gytochrome othE? ’hemoprotein systems. S. Fleischer Packégv-eds. Acaam mic 30. Burk, R. F. 1983. Glutathione-dependent protection by rat liver microsomal protein against lipid peroxidation. Biochim. 31. Burk, R. F. and R. .A. Lawrence. 1978. Non-selenium-dependent glutathione peroxidase. In Functions .9: Glutathione in Liver and Kidney. H. Sies and A. wendel, eds. Springer-Verlag, New York. pp. 114-119. 32. Cha, Y-N. 1978. Inducibility of the hepatic drug-metabolizing capacity of mice infected with Schistosoma mansoni. Am. J. Trop. Med. Hyg . 27: 1181-1187. 33. Cha, Y-N. and Edwards, R. 1976. Effect of Schistosoma mansoni infection on the hepatic drug-metabolizing capacity of mice. J. Pfiarmac. Exp. Therap. 199: 432-440. 34. Chance, B. Sies, H. and Boveris, A. 1979. Hydroperoxide metabolism in mammalian organs. Physiol. Rev. 59: 527-605. 35. Chappell, L. H. 1974. Methionine uptake by larval and adult Schistosoma mansoni. Int. J. Parasitol. 4: 361-369. 36. Chow, C. K. and Tappel, A. L. 1974. An enzymatic mechanism against lipid peroxidation damage to lungs of ozone exposed rats. Lipids 7: 518-524. 37. Coles, G. C. 1973. Enzyme levels in cercariae and adult Schistosoma mansoni. Int. J. Parasitol. 3: 505-510. 38 . Cornford , E. M. and Oldendorf , W. H. 1979. Transintegumental uptake of metabolic substrates in male and female Schistosoma mansoni. J. Parasitol. 65: 357—363. 39. Dartigues, B., Cambar, J., Trebaul, C., Brelivet, J. and Guglielmetti, R. 1980. Proprietes diuretiques de derives des dithiole-thiones. Recherche d'une relation structure—activite. Chim. Therap. 15: 405-412. 65 40. Davis, J. S., Balinsky, J. B. and Harington, J. S. 1973. Assay, purification, prOperties and mechanism of action Of Y-glutamylcysteine synthetase from the liver of the rat and Xenopus laevis. Biochem. J. 133: 667-678. 41. Douch, P. G. C. and Blair, 8. S. B. 1975. The metabolism of foriegn compounds in the cestode, Monezia e ansa and the nematode Ascaris lumbricoides var suum. Xenobiotica 5: 279-292. 42. Douch, P. G. C. and Buchanan, L. L. 1978. Glutathione conjugation of some xenobiotics by Ascaris suum and Moniezia expansa. Xenobiotica 8: 171-176. 43. Douch, P. G. C. and Gahagan, H. M. 1977. The metabolism of niclosamide and related compounds by Moniezia expgnsa, Ascaris lumpgicoides var suum and mouse- and sheep-liver enzymes. XenObiotIca 7: 301-307. 44. Ellman, G. L. 1959. Tissue sulfhydryl groups. Arch. Biochem. BiOphys. 82: 70-77. 45. Fetterer, R. H., Pax, R. A. and Bennett, J. L. 1977. Schistosoma mansoni: Direct method fOr simultaneous recording Of electrical and motor activity. Exp. Parasitol. 43: 286-294. 46. Fioravanti, C. F. 1982. Mitochondrial malate dehydrogenase, decarboxylating ("malic" enzyme) and transhydrogenase activities of adult Hymenolepis microstoma (Cestoda). J. Parasitol. 68: 213-220. 47. Fioravanti, C. F. and Saz, H. J.. 1976. Pyridine nucleotide tranhydrogenases of parasitic helminths. Arch. Bioch. Biophys. 175: 21-30. 48. Flohe, L. and Grunzler, W; A. 1976. Glutathione-dependent enzymatic oxidoreduction reactions. In Glutathione: Metabolism and Function. I. M. Arias and W: B. Jakoby, eds. Raven Press, New York. pp. 17-34. 49. Frischer, H. and Ahmad, T. 1977. Severe generalized glutathione reductase deficiency after antitumor chemotherapy with BCNU (l,3-bis[chloroethyl]-l— nitrosourea). J. Lab. Clin. Mad. 89: 1080-1091. 50. Gentilini, M. Duflo, B., Richard-Lenoble, D., Brucker, G. Danis, M. Neil, G. and Meunier, Y. 1980. Assessment of 35972 RP (oltipraz) a new antischistosomal drug against Schistosoma haematobium, Schistosoma mansoni, and Schistosoma intercalatum. Acta Trop. 37: 271-274. 66 51. Gilbert, H. F. 1982. Biological disulfides: The third messenger? Modulation of phosphofructokinase activity' by thiol/disulfide exchange. J. Biol. Chem. 257: 12086-12091. 52. Gillette, J. R., Mitchell, J. R. and Brodie, B. B. 1974. Biochemical mechanisms of drug toxicity. Ann. Rev. Pharmacol. 14: 271-288. 53. Goodman, J. and Hochstein, P. 1977. Generation of free radicals and lipid peroxidation by redox cycling of adriamycin and daunomycin. Biochem. biOphys. Res. Comm. 77: 797-803. 54. Grant, C. T. and Senft, A. ‘W. 1971. Schistosome proteolytic enzyme. comp. Biochem. Physiol. 38: 663-678. 55. Griffith, O. ‘W. 1980. Determination of glutathione and glutathione disulfide using glutathione reductase and 2-vinylpyridine. Anal. Biochem. 106: 207-212. 56. Griffith, O. ‘W. 1981. Depletion of glutathione by inhibition of biosynthesis. Methods Enzymol. 77: 59-63. 57. Griffith, O. ‘W. 1982. Mechanism of action, metabolism, and toxicity of buthionine sulfoximine and its higher homologs, potent inhibitors of glutathione synthisis. J. Biol. Chem. 257: 13704-13712. 58. Griffith, O. ‘W. 1981. Glutathione turnover in human erythrocytes: Inhibition by buthionine sulfoxime and incorporation Of glycine by exchange. J. Biol. Chem. 256: 4900-4904. 59. Griffith, O. W; and Meister, A. 1977. Selective inhibition Of ‘Y-glutamyl-cycle enzymes by substrate analogs. Proc. Nat. Acad. Sci. USA 74: 3330-3334. 60. Griffith, O. W. and Meister, A. 1979. Potent and specific inhibition of glutathione synthesis by buthionine sulfoximine (S-n—butyl homocysteine sulfoximine). J. Biol. Chem. 255: 7558-7560. 61. Griffith, O. W. and Meister, A. 1979. Translocation Of intracellular glutathione to membrane-bound Y-glutamyl transpeptidase as a discrete step in the Y-glutamyl cycle. Glutathionuria after inhibition of transpeptidase. Proc. Nat. Acad. Sci. USA 76: 268-272. 62. Griffith, O. ‘W. and Meister, A. 1979. Glutathione: Interorgan translocation, turnover, and metabolism. Proc. Natl. Acad. Sci. USA 76: 5606-5610. 67 63. Griffith, O. ‘W. and Meister, A.. 1980. Excretion of cysteine and Y-glutamylcysteine moieties in human and experimental animal Y—glutamyl transpeptidase deficiency. Proc. Nat. Acad. Sci. USA 77: 3384-3387. 64. Habig,‘W. H., Pabst, M. J. and Jacoby, ‘W. B. 1974. Glutathione S-Transferases: The first enzymatic step in mercapturic acid fOrmation. J. Biol. Chem. 249: 7130-7139. 65. Hafeman, D. C., Sunde, R. A. and Hoekstra, W; G. 1974. Effect of dietary selenium on erythrocyte and liver glutathione peroxidase in the rat. J. Nutr. 104: 580-587. 66. Hahn, R and Oberrauch, W& 1978. unidirectional transport of reduced glutathione in rat liver and its metabolization in the extracellular space. In Glutathione: Metabolism and Function. I. M. Arias and W: B. Jakoby, eds. Raven Press, New York. pp. 32-40. 67. Harris, J. W; and Biaglow, J. E. 1972. Non-specific reactions of the glutathione oxidant "diamide" with mammalian cells. Biochem. Biophys. Res. Cbmm. 46: 1743-1749. 68. Higashi, T., Tateishi, N., Naruse, A. and Sakamoto, Y. 1977. A novel physiological role of liver glutathione as a reservoir of L-cysteine. J. Biochem. 82: 117-124. 69. Hoffman, D. B. and Bennett, J. L. 1979. IV. Drug development strategy. Clark Foundation Advisory Report. 70. Hogberg, J. and Kristoferson, A. 1977. A. correlation between glutathione levels and cellular damage in isolated hepatocytes. Eur. J. Biochem. 74: 77-82. 71. Isseroff, H. and Levy, M. 1972. Amino acid transport in Schistosoma mansoni. Am. Zool. 12: 681. 72. Ito, K., Vakagawa, J., Minkuohi, C. and Fukase, M. 1971. A clinical evaluation of serum monoamine oxidase, with special reference to hepatic fibrosis. Digestion 4: 49-58. 73. Jacob, H. S. and Jandl, J. H. 1962. Effects of sulfhydryl inhibition on red blood cells. I. Mechanism of hemolysis. 74. Jacoby, ‘W. B. 1978. The glutathione transferases in detoxification. in Functions Of Glutathione in Liver and Kidne (Sies, H. and Wendel, A. Eds.) spEInger-Verlag,'_§erIin,'_H§ide rg, New York. pp 157-163. 75. Jacoby, W; B. and Habig, ‘W. H. 1980. Glutathione transferases. in Enzymatic Basis gleetoxication, VOl. II. (W: B. Jacoby, ed.) Academic Press, Inc., New York. pp 63-94. 68 76. Jackson, R. C. 1969. Studies in the enzymology of glutathione metabolism in human erythrocytes. Biochem. J. 111: 309-315. 77. James, R. C. and Harbison, R. D. 1982. Hepatic glutathione and hepatotoxicity: Effects of cytochrome P-450 complexing compounds SKF 525-A, L-a acetylmethadol (LAAM), norLAAM, and piperonyl butoxide. Biochem. Pharmac. 31: 1829-1835. 78. JOcelyn, P. C. 1978. The reduction of diamide by rat liver mitochondria and the role of glutathione. Biochem. J. 176: 649-664. 79. JOlles, G. 1984. Pharmacokinetics, metabolism and mechanism of action of oltipraz in animals and man. WHO Scientific WOrking Group, Geneva. 80. JOrdan, P. 1977. Schistosomiasis - Research to control. Am. J. Trop. Med. Hyg. 26: 877-885. 81. Kaplowitz, N. 1981. The importance and regulation of hepatic glutathione. Yale J. Biol. Med. 54: 497-502. 82. Keeling, P. L. and Smith, L. L. 1982. Relevance of NADPH depletion and mixed disulphide fOrmation in rat lung to the mechanism of cell damage fOllowing paraquat administration. Biochem. Pharmacol. 31: 3243-3249. 83. Ketterer, B. 1982. The role of nonenzymatic reactions Of glutathione in xenobiotic metabolism. Drug metabolism reviews. 13: 161-187. 84. Ketterer, B., Cbles, B. and Meyer, D. J. 1983. The role of glutathione in detoxication. Envir. Health Perspect. 49: 59-69. 85. Kohler, P. and Saz, H. J. 1976. Demonstration and possible function of NADH:NAD+ transhydrogenase from Ascaris muscle mitochondria. J. Biol. Chem. 251: 2217-2225. 86. Kbsower, E. M. and Kosower, N. S. 1969. Lest I forget thee, glutathione ... Nature 224: 117-120. 87. KOsower, N. S. and Kosower, E. M. 1978. The glutathione status of cells. Intern. Rev. Cyt. 54: 109-160. 88. Kosower, N. S., Kosower, E. M. and Wertheim, B. 1969. Diamide, a new reagent for the intracellular oxidation Of glutathione to the disulfide. Biochem. Biophys. Res. Cbmm. 37: 593-596. 89. Kbsower, N. S., Zipser, Y. and Faitin, Z. 1982. Membrane thiol-disulfide status in glucose-6-phosphate dehydrogenase deficient red cells: Relationship to cellular glutathione. Biochim. biophys. Acta 691: 345-352. ' 90. Kurelec, B. and Rijavec, M. 1976. Occurence of Y-glutamyl cycle in some parasitic helminths. In Biochemistpy of Parasites and Host-Parasite Relationships. H. van den '_EOSsche, ed. Elsevier/North-Holland BiOmedical Press,, Amsterdam. pp. 101-107. 91. Lawrence, R. A. and Burk, R. F. 1976. Glutathione peroxidase activity in selenium-deficient rat liver. Biochem. Biophys. Res. Cbmm. 71: 952-958. 92. Leroy, J. P., Barreau, M., Ootrel, C., Jeanmart, C., Messer, M. and Benazet, F. 1978. Laboratory studies of 35972 RP, a new schistosomicidal compound. Current Chemotherapy 1: 148-150. 93. Little, C. and O'Brien, P. J. 1968. An intracellular GSH-peroxidase with a lipid peroxide substrate. Biochem. Biophys. Res. Cbmm. 31: 145-150. 94. Lowry. O. H., Rosebrough, N. J., Farr, A. L. and Randall, R. J. 1951. Protein measurement with the Folin phenol reagent. J. Biol. Chem. 193: 265-275. 95. Meister, A. 1973. On the enzymology of amino acid transport. Science 180: 33-39. 96. Meister, A. 1974. Biosynthesis and utilization of glutathione; the ‘Y-glutamyl cycle and its function in amino acid transport. in Glutathione (L. Flohe, H. Ch. Benohr, H. Sies, H. D. Waller and A. Wendel, eds) Academic Press, Inc., New York. pp 56-67. 97. Meister, A. 1978. Current status of the Y-glutamyl cycle. In Functions ‘9§_ Glutathione 119' Liver and Kidney. H. Sies and A. wendel, eds. Springer-verlag, New'YOrk. pp. 43-59. 98. Meister, A. and Anderson, M. E. 1983. Glutathione. Ann. Rev. Biochem. 52: 711- 760. 99. Meister, A. and Tate, S. S. 1976. Glutathione and related Y-glutamyl compounds: Biosynthesis and utilization. Ann. Rev. Biochem. 45: 559-604. 100. Mills, G. C. 1959. The purification and properties of glutathione peroxidase of erythrocytes. J. Biol. Chem. 234: 502-506. 101. Moldeus, P., Onmstad, K. and Reed, D. J. 1981. TUrnover of cellular glutathione in isolated rat-kidney cells: Role Of cystine and methionine. Eur. J. Bioch. 116: 13-16. 102. Moore, D. V. and Sandground, J. H. 1956. The relative egg producing capacity of Schistosoma mansoni and Schistosoma japgnicum. Am. J. Trop. Med. Hyg. 5: 831-840. 70 103. Morello, A., Repetto, Y. and Atias, A. 1982. Characterization of glutathione S—transferase activity in Echinococcus granulosus. Comp. Biochem. Physiol. 728: 449-452. 104. Moron, M. S., Depierre, J. ‘W. and Mannervik, B. 1979. Levels of glutathione, glutathione reductase and glutathione S-transferase activities in rat lung and liver. Biochim. biophys. Acta, 582: 67-78. 105. Nishiki, K., Jamieson, D., Oshino, N. and Chance, B. 1976. Oxygen toxicity in the perfused rat liver and lung under hyperbaric conditions. Biochem. J. 160: 343-355. 106. Noelle, R. J. and Lawrence, D. A. 1981. Modulation of T-cell function. II. Chemical basis for the involvement of cell surface thiol-reactive sites in control Of Thcell proliferation. Cell. Immunol. 60: 453-469. 107. Cffermann, M. K., McKay, M. J., Marsh, M. ‘W. and Bond, J. S. 1984. Glutathione disulfide inactivates, destabilizes and enhances proteolytic susceptibility of fructose-l,6-bis-phosphate aldolase. J. Biol. Chem. 259: 8886-8891. 108. Orlowski, M. and Meister, A. 1970. Y-glutamyl transpeptidase (hog kidney) In Methods 12_Enzymology 17A. H. Tabor and C. W. Tabor, eds. Academic Press, New YOrk and London. pp. 883-889. 109. Ormstad, K., Jones, D. P. and Orrenius, S. 1980. Characteristics of glutathione biosynthesis by freshly isolated rat kidney cells. J. Biol. Chem. 255: 75-81. 110. Pang, K.-Y., Y., Braswell, L. M., Chang, L., Sommer, T. J. and K. ‘W. Miller. 1980. The perturbation of lipid bilayers by general anesthetics: A quantitative test of the disordered lipid hypothesis. Mol. Pharmacol. 18: 84-90. 111. Pieron, R., Lesobre, B., Marfart, Y. and Lancastre, F. 1980. Effets de l'oltipraz en traitment bref dans la bilharziose. (en comparaison avec le niridazole). Med. Trop. 40: 302-311. 112. Pillon, D. J., Moree, L., Rocha, H., Pashley, D. H., Medicino, J. and Leibach, F. H. 1977. The rple Of glutathione in renal cortical tissue. Eff ct of diamide on Na and GSSG levels, amino acid transport and Na -K -ATPase activity. Mol. Cell. Biochem. 18: 109-115. 113. Prohaska, J. R. 1980. The glutathione peroxidase activity of glutathione S-transferases. Biochim. biophys. Acta 611: 87-98. 71 114. Reed, D. J, Babson, J. R., Beatly, P. W., Brodie, A. E., Ellis, W. W. and Potter, D. W. 1980. High—performance liquid chromatography analysis of nanomole levels of glutathione, glutathione disulfide, and related thiols and disulfides. Anal Biochem. 106: 55-62. 115. Reed, D. J. and Beatty, P. ‘W. 1978. The role of cystathionine pathway in glutathione regulation by isolated hepatocytes. In Functions 9f Glutathione '13 Liver and Kidney. H. Sies and A. wendel, eds. Springer-verlag, New York. pp. 13-21. 116. Reed, D. J. and Orrenius, S. 1977. The role of methionine in glutathione biosynthesis by isolated hepatocytes. Biochem. Biophys. Res. Comm. 77: 1257-1264. 117. Richman, P. G. and Meister, A. 1975. Regulation of Y—glutamyl—cysteine synthetase by nonallosteric feedback inhibition by glutathione. J. Biol. Chem. 250: 1422-1426. 118. Robinson, C. H., Bueding, E. and Fisher, J. 1970. Relationship between structure, conformation, and antischistosomal activity of nitroheterocyclic compounds. Mol. Pharmacol. 6: 604-616. 119. Schwarz, K. 1965. Role of vitamin E., selenium, and related factors in experimental nutritional liver disease. Fed. Proc. 24: 58-67. 120. Seed, J., unpublished studies 121. Seaman, P. 1972. The membrane actions of anesthetics and tranquilizers. Pharmacol. Rev. 24: 583—655. 122. Semeyn, D. R., Pax, R. A. and Bennett, J. L. 1982. Surface electrical activity from Schistosoma mansoni: A sensitive measure of drug action. J. Parasitol. 68: 353-362. 123. Senft, A. 1963. (bservations on amino acid metabolism of Schistosoma mansoni in a chemically defined media. Ann. N. Y. Acad. Sci. 113: 272—288. 124. Sheetz, M. P. 1980. in Genetic disorders of glutathione and sulfer amino-acid metabolism (Schulman, J. D., moderator) Ann. Intern. Med. 93: 341-343. 125. Siegers, C. -P., Hubscher, W. and Younes, M. 1982. G1utathione-S-transferase and- GSH-peroxidase activities during the state of GSH-depletion leading to lipid peroxidation in rat liver. Res. Comm. Chem. Pathol. Pharmac. 37: 163-169. 72 126. Szasz, G. 1969. A kinetic photometric method from serum Y-glutamyl transpeptidase. Clin. Chem. 15: 124-136. 127. Tappel, M. E., Chaudiere, J. and Tappel, A. L. 1982. Glutathione peroxidase activities of animal tissues. Comp. Biochem. Physiol. 738: 945-949. 128. Tateishi, N., Higashi, T., Shinya, S., Naruse, A. and Sakamoto, Y. 1974. Studies on the regeneration of glutathione level in rat liver. J. Biochem. 75: 93-103. 129. Thompson, D. P., Pax, R. A. and Bennett, J. L. 1982. Microelectrode studies on the tegunental and subtegunental compartments of male Schistosoma mansoni: An analysis of electrophysiological properties. ParaSitology 85: 163-178. 130. Tietze, F. 1969. Enzymic method fOr quantitative determination of nanogram amounts of total and oxidized glutathione: Applications to mammalian blood and other tissues. Anal. Biochem. 27: 502-522. 131. uglem, G. L. and Read, C. P. 1975. Sugar transport and metabolism in Schistosoma mansoni. J. Parasitol. 61: 390-397. 132. warren, K. S. 1970. Suppression of hepatosplenic schistosomiasis mansoni in mice by nicarbazin, a drug that inhibits egg production by schistosomes. J. Inf. Dis. 121: 514-521. 133. warren, K. S. 1978. Hepatosplenic schistosomiasis: a great neglected disease of the liver. Gut 19: 572-577. 134. warren, K. S. 1978. The pathology, pathobiology and pathogenesis of schistosomiasis. Nature 273: 609-612. 135. Wfinslow, D. J. 1967. HistOpathology of schistosomiasis. in Bilharziasis (F. K. Mostofi, ed). Springer-verlag, New York. pp. 136. Y0unes, M. and Siegers, C. -P. 1981. Mechanistic aspects of enhanced lipid peroxidation fOllowing glutathione depletion in_vivo. Chem. -Biol. Interactions 34: 257-266. ”'il’iifimglljlflfijlfillififiujifllu‘tflffllflm‘“