antioxidant defense mechanisms in parasitic protozoa

20
Critical Reviews in Microbiology. 22(4):295-3 14 (1 996) Antioxidant Defense Mechanisms in Parasitic Protozoa Rajeev K. Mehlotra Division of Geographic Medicine, School of Medicine, W123, Case Western Reserve University, 2109 Adelbert Road, Cleveland, OH 441 06-4983 ABSTRACT: Many of the parasitic protozoa, such as Entamoeba histolytica, Giardia, Trypa- nosoma, Leishmania, and Plasmodium, are considered to be anaerobes because they can be grown in vitro only under conditions of reduced oxygen tension. However, these parasitic protozoa have been found to be aerotolerant or microaerophilic, and also to consume oxygen to a certain extent. Furthermore, these organisms are highly susceptible to exogenous reactive oxygen species, such as hydrogen peroxide. They must, therefore, detoxify both oxygen and free radical products of enzymatic reactions. However, they lack some or all of the usual antioxidant defense mechanisms present in aerobic or other aerotolerant cells, such as catalase, superoxide dismutase, reduced glutathione, and the glutathione-recycling enzymes glutathione peroxidase and glutathione reductase. Instead, they possess alternative mechanisms for detoxification simi- lar to those known to exist in certain prokaryotes. Although the functional aspects of these alternative mechanisms are yet to be understood completely, they could provide new insights into the biochemical peculiarities of these enigmatic pathogens. KEY WORDS: antioxidants, parasites, protozoa, Entamoeba histolytica, Giardia, Trypano- soma, Leishmania, Plasmodium. 1. INTRODUCTION Aerobic organisms produce toxic oxy- gen derivatives by the reduction of molecu- lar oxygen during aerobic metabolism. The most common free-radical products are the superoxide anion (O;), hydrogen peroxide (H,O,), and their breakdown or metabolic products. Accordingly, they possess elabo- rate defense mechanisms to detoxify the (toxic) products of oxygen reduction. Super- oxide dismutase (SOD) catalyzes the dismutation of 0; to H,O, + 0,. H,O, is converted to H,O + 0, by catalase, peroxi- dase, and glutathione peroxidase, which uses glutathione (GSH) as the reducing agent. Two major types of SOD carry out the dismutation of 0; in different organisms and within different organelles and cellular com- partments. These SODSare characterized by the presence of metal ions (either Mn/Fe or Cu and Zn) at the active site of the enzyme, and their sensitivity to cyanide, azide, and H20,. Bacteria contain Mn/Fe- and/or Cu.Zn- SOD, while virtually all eukaryotes contain both types of In most eukaryotes, Mn-SOD activity is restricted to the mito- chondria and chloroplast; the enzyme is in- sensitive to cyanide and H,O,. Fe-SOD has been found in plants and several free-living and parasitic protozoa; the enzyme is inhib- ited by H,O,, but not by cyanide. Cytosolic sources of 0; are handled in most higher eukaryotes by Cu.Zn-SOD. Recently, the enzyme has also been described from the periplasm of a number of This 1040-84 1 x/96/$.50 Q 1996 by CRC Press, Inc. 295 Critical Reviews in Microbiology Downloaded from informahealthcare.com by University of British Columbia on 04/29/13 For personal use only.

Upload: rajeev-k

Post on 09-Dec-2016

212 views

Category:

Documents


0 download

TRANSCRIPT

  • Critical Reviews in Microbiology. 22(4):295-3 14 ( 1 996)

    Antioxidant Defense Mechanisms in Parasitic Protozoa

    Rajeev K. Mehlotra Division of Geographic Medicine, School of Medicine, W123, Case Western Reserve University, 21 09 Adelbert Road, Cleveland, OH 441 06-4983

    ABSTRACT: Many of the parasitic protozoa, such as Entamoeba histolytica, Giardia, Trypa- nosoma, Leishmania, and Plasmodium, are considered to be anaerobes because they can be grown in vitro only under conditions of reduced oxygen tension. However, these parasitic protozoa have been found to be aerotolerant or microaerophilic, and also to consume oxygen to a certain extent. Furthermore, these organisms are highly susceptible to exogenous reactive oxygen species, such as hydrogen peroxide. They must, therefore, detoxify both oxygen and free radical products of enzymatic reactions. However, they lack some or all of the usual antioxidant defense mechanisms present in aerobic or other aerotolerant cells, such as catalase, superoxide dismutase, reduced glutathione, and the glutathione-recycling enzymes glutathione peroxidase and glutathione reductase. Instead, they possess alternative mechanisms for detoxification simi- lar to those known to exist in certain prokaryotes. Although the functional aspects of these alternative mechanisms are yet to be understood completely, they could provide new insights into the biochemical peculiarities of these enigmatic pathogens.

    KEY WORDS: antioxidants, parasites, protozoa, Entamoeba histolytica, Giardia, Trypano- soma, Leishmania, Plasmodium.

    1. INTRODUCTION

    Aerobic organisms produce toxic oxy- gen derivatives by the reduction of molecu- lar oxygen during aerobic metabolism. The most common free-radical products are the superoxide anion (O;), hydrogen peroxide (H,O,), and their breakdown or metabolic products. Accordingly, they possess elabo- rate defense mechanisms to detoxify the (toxic) products of oxygen reduction. Super- oxide dismutase (SOD) catalyzes the dismutation of 0; to H,O, + 0,. H,O, is converted to H,O + 0, by catalase, peroxi- dase, and glutathione peroxidase, which uses glutathione (GSH) as the reducing agent. Two major types of SOD carry out the dismutation of 0; in different organisms and

    within different organelles and cellular com- partments. These SODS are characterized by the presence of metal ions (either Mn/Fe or Cu and Zn) at the active site of the enzyme, and their sensitivity to cyanide, azide, and H20,. Bacteria contain Mn/Fe- and/or Cu.Zn- SOD, while virtually all eukaryotes contain both types of In most eukaryotes, Mn-SOD activity is restricted to the mito- chondria and chloroplast; the enzyme is in- sensitive to cyanide and H,O,. Fe-SOD has been found in plants and several free-living and parasitic protozoa; the enzyme is inhib- ited by H,O,, but not by cyanide. Cytosolic sources of 0; are handled in most higher eukaryotes by Cu.Zn-SOD. Recently, the enzyme has also been described from the periplasm of a number of This

    1040-84 1 x/96/$.50 Q 1996 by CRC Press, Inc.

    295

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.

  • form of SOD is inhibited by both cyanide and H,O,. All of the SODS are inhibited by azide.

    Various parasitic protozoa, such as En- tamoeba histolytica, Giardia, trichomonads, Trypanosoma, Leishmania, and Plasmodium, have been shown to be anaerobic in their cultivation requirements. However, metaboli- cally, they have been found to be micro- aerobic or microaerophilic. They have also been found to produce H,02 as a product of their metabolism in certain cases, and to be highly sensitive to exogenous H,O,. More- over, these parasites, especially the intracel-

    lular ones, are naturally exposed to the toxic oxygen metabolites generated by effector immune cells of the host. However, many of them have been reported to lack, or to be extremely deficient in, enzyme systems nec- essary for the detoxification of H202 (i.e., catalase, glutathione peroxidase) (Table 1). Instead, they possess alternative mechanisms for the detoxification, which have been well characterized from certain prokaryotes (Table 2). The purpose of this review is to provide a broad overview of the strategies, both con- ventional and alternative, of these parasitic protozoa for the detoxification of oxygen

    TABLE 1 Conventional Antioxidant Defense Mechanisms in Various Protozoan Parasites

    Parasite

    Entamoeba histolytica

    Giardia Trypanosoma Leishmania (A) Plasmodium bergheP

    P. falciparum Trichomonas vaginalis

    Tritrichomonas foetus

    Reduced Superoxide glutathionel

    Catalase dlsmutase trypanothione

    + + + - + -?

    - + +

    Glutathionel trypanothlone

    reductase

    - + + +

    + -?

    -?

    Glutathionel trypanothione

    peroxldase

    - +/- +/- +

    + -?

    -?

    Note: +, present: -, absent; +I-, uncertain: -?, not likely to be present; (A) = Amastigotes. lntraerythrocytic stages.

    TABLE 2 Alternative Antioxidant Defense Mechanisms in Certain Protozoan Parasites

    NAD(P)H Ascorbate Parasite Cystelne NAD(P)H oxldase peroxldase peroxldase

    Entamoeba + + histolytica

    Giardia + + Trypanosoma - Trichomonads + +

    +

    ND ND

    + ND + +!-

    ND + Note: +, present; -, absent; +I-, uncertain: ND, not demonstrated.

    296

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.

  • metabolites relevant to their survival in their mammalian host. Recent developments and future directions for research in this area are also outlined.

    I I . ENTAMUEBA HISTULYTICA

    The normal environment of the tropho- zoites (feeding stages) of E. histolytica is essentially anaerobic, and maintenance of low redox potential is obligatory to its opti- mal growth in vitro. To maintain a reduced potential in axenic cultures, investigators have used a variety of reducing agents. A combination of L-cysteine (0.1%) and ascor- bic acid (0.02%) as the reducing agent was used in the axenic TP-S-1 and TYI-S-33 (or BI-S-33) media.5p6 Good growth of the amoe- bae in these media was also achieved using L-cysteine (0.2%) without ascorbic a ~ i d , ~ . ~ L-ascorbic acid, D-cysteine, L-cystine, or dithiothreitol: or sodium thioglycollate.'O We have found that GSH (0.2%) supported as good a growth of E. histolytica as the com- bination of L-cysteine and ascorbic acid, or L-cysteine (0.2%) in BI-S-33 medium." Moreover, 0.25% GSH supported consider- ably higher growth of amoebae, similar to that obtained with 0.3% L-cysteine."

    E. histolytica is an anaerobe. However, the pathogen has also been shown to be oxy- gen tolerant and to consume oxygen under certain conditions.12 It can tolerate up to 5% oxygen in the gas phase and is able to detoxlfy the products of oxygen reduction in the medium.I3 Although E. histolytica is prima- rily a colonic pathogen, it also invades other organs, mainly the liver. Amoebic liver ab- scess results from pathogenic trophozoites moving from the colon to the liver via the portal circulation. Thus, neither the tissues invaded by the amoebae nor the media used for their cultivation are notably deficient in oxygen. Curiously, E. histolytica lacks cata- lase,12J4 peroxidase,14 and enzymes of glu-

    tathione me tabo l i~m,~~ but Fe-SOD has been found in this p a t h ~ g e n . ' ~ . ~ ~ Hence, if toxic 0; is formed by the partial reduction of oxy- gen in a variety of aerobic enzymatic oxida- tions, it would be removed with the forma- tion of H,02. How H202 would be detoxified in the absence of catalase is not known. H20, has been shown to be amoebicidal in v i m using an enzymatic H,02-generating systemI7 or in solution.18 It was also found that the addition of catalase to the axenic medium could protect the trophozoites from the le- thal effects of H,O2.I8

    A point of interest is that the concentra- tion of reducing agent required in the axenic medium (e.g., 0.1% w/v cysteine) is consid- erably higher than that required for the growth of anaerobic bacteria, in which 0.02 to 0.05% cysteine is adequate and higher concentra- tions are usually toxic.19 This suggests cer- tain other roles for these reducing agents for the organism, in addition to the maintenance of low redox potential to facilitate growth. Both L-cysteine and L-cystine were found to protect E. histolytica trophozoites from the lethal effects of increased PO, in TYI-S-33 medium.'O The mechanism of protection by L-cystine is obscure, but must be unusual because this amino acid lacks a thiol reduc- ing group. We have proposed that a possible factor effective in detoxifying the H20, might be the availability of "excess" sulfhy- dry1 (-SH) groups.21 This is supported by the observation of Band and Cirrito13 that unin- oculated culture medium containing 0.1% cysteine, exposed to 5% oxygen for 24 h, would support normal growth of amoebae on subsequent inoculation. However, expo- sure for 72 h was unfavorable for the organ- ism, possibly due to the accumulation of toxic products of oxygen reduction in excess of the ability of the organism to deal with them. The toxicity of the medium could be reversed by adding fresh cysteine, indicating that -SH compounds can detoxify products of oxygen reduction. Cysteine, freshly added

    297

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.

  • to the axenic medium,, was found to be rap- idly oxidized under conditions used for cul- t ~ r e , ~ and the fact that GSH normally oxidizes less rapidly than cysteine might be important for its better growth-supporting ability. It is also important to mention here that E. histolytica, grown on normal TYI-S-33 me- dium, has been found to contain significant amounts of glutathione, but only 1/3 of the total was present in the reduced form. How- ever, in glutathione-depleted medium, cys- teine was the main thiol component in E. histolytica and was present largely in the thiol

    Thus, the reducing agents (thiols) may be utilized for detoxification in E. histolytica, which otherwise has a limited ability to detoxify the products of oxygen reduction.

    Reed et al., reported the isolation of an E. histolytica cDNA clone encoding a cys- teine-rich 29-kDa protein. The function of the protein is unknown and its localization is not well defined. A database analysis re- vealed substantial sequence homology of the 29-kDa protein to a class of polypeptides found in prokaryotic organisms that may be involved in the inactivation of H202.23 Re- cently, another gene has been identified in E. histolytica encoding a protein (34-kDa) ho- mologous to prokaryotic disulfide oxi- doreductases, and it has been suggested that the 29- and 34-kDa proteins represent the two subunits of the active enzyme.24 A more detailed analysis of the protein may help to determine its role in the protection of amoe- bae against toxic oxygen metabolites. Bruchhaus and TannichZ5 studied the regula- tion of SOD expression in four E. histolytica isolates under different culture conditions. They found that incubation of amoebae in the presence of an 0,-generating system re- sulted in an increase in SOD activity be- tween 3.2- and 4.7-fold, as well as an in- crease in Fe-SOD protein concentration between 2.7- and 5.5-fold. Incubation of amoebae in the presence of a ferrous iron chelator resulted in an increase in SOD

    activity between 1.7- and 5.3-fold and in Fe- SOD protein concentration between 2.8- and 7.2-fold. Based on these observations, the authors suggested the possibility that such a regulation of SOD may contribute to protec- tion from toxic oxygen metabolites during tissue invasion by E. histolytica.

    111. GIARDIA

    Giardia is another anaerobic, amito- chondrial protozoan which has been culti- vated axenically in modified TP-S- 1 or TYI- S-33 media with reduced oxygen tension, using L-cysteine and ascorbic acid as the reducing However, growth re- sponses of G. lamblia to reducing agents have been found to be quite different from those observed for E. hi~tolytica.~ In TYI-S- 33 medium, the requirement for reducing agents was found to be quite specific. Only L-cysteine supported the maximum growth of the organism, whereas in TP-S- 1 medium GSH, N-acetyl L-cysteine and mercapto- propionyl glycine also supported 60 to 70% of the maximum growth. L-Ascorbic acid, D- cysteine, and L-cystine were totally ineffec- tive in both the media. However, a combina- tion of L-cystine and ascorbic acid yielded limited growth in TP-S-1 medium.

    Giardia does respire and consume oxy- gen, in that oxygen is actively removed at low partial pressures from culture vessel^.^^^^^ However, the parasite lacks SOD, catalase, peroxidase, and GSH.30.31 L-Cysteine, as in E. histolytica, was found to protect G. lamblia trophozoites under high PO,.~O Although ascorbic acid did not support the growth of the organism, it also protected the tropho- zoites under high PO,, as did L-cysteine. In another cysteine as well as GSH were found to protect G. lamblia trophozo- ites from thiol-blocking reactants, indicating a role for the reducing agents for protection of crucial thiol groups.

    298

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.

  • Brown et al.30 have found cysteine to be the major low-molecular-weight thiol in Giardia. Low levels of sulfite, thioglycollic acid, and coenzyme A were also observed, while GSH and its intermediates were not detectable. They have also shown that the microaerophilic, amitochondrial parasite Tritrichomonas foetus lacked GSH, but con- tained high levels of cysteine. Lujan and N a ~ h ~ ~ studied the uptake and metabolism of cysteine by G. lamblia trophozoites. They concluded that L-cysteine is an essential growth factor for Giardia because the para- site (1) cannot take up L-cystine and (2) cannot synthesize cysteine de novo. They have also suggested that the role of cysteine in Giardia may be to protect the organism from the lethal effects of oxygen, as in E. histolytica and Trichomonas vaginalis. 34 Recently, Brown et aL3* found that Giardia contains an alternative mechanism of oxy- gen detoxification to those seen in higher eukaryotes. High-level NADH-dependent oxidase and low-level NADH-dependent peroxidase activities were detected, similar to anaerobic bacteria. E. histolytica, Tritrichomonas foetus, and Trichomonas vaginalis were also found to contain NADH oxidase activity, confirming earlier observa- t i o n ~ . ~ ~ . ~ ~ However, the authors were unaware of any other reports of an NADH peroxidase being demonstrated in the trichomonads and Entumoeba. Taken together, these enzyme activities, complemented by the high con- tents of endogenous cysteine and thiogly- collic acid (a free-radical trap), seem to pro- vide an effective detoxification mechanism in Giardia.

    IV. TRYPANOSOMA

    Long-term cultivation of pathogenic African trypanosomes in vitro is now well established. To grow trypanosomes axeni- cally, the addition of reducing agents such as

    cysteine, 2-mercaptoethanol, or monothio glycerol to the medium was found to bt necessary for continuous c ~ l t i v a t i o n . ~ ~ . ~ ~ Hamm et al.39 found that monothioglycero was the optimal reducing agent for the dif. ferentiation of Trypanosoma brucei trypo- mastigotes in axenic culture because it sup. ported constant growth rates in all subcultures and was more stable than either cysteine 01 2-mercaptoethanol. Using a cystine-free. minimum essential medium for the axenic cultivation of T. brucei, Duszenko et aL4 found that cell growth was only observed if L-cysteine was either directly added to the medium or was reduced from cystine by the action of reducing agents (monothioglycerol or 2-mercaptoethanol). However, neither reducing. agents alone nor D-cysteine sup- ported cell growth. The need for cysteine was obligatory, despite the fact that methion- ine (a precursor of cysteine biosynthesis) was a constituent of the growth medium. Moreover, T. brucei has been found to effec- tively transport only cysteine and not cys- tine.38 These observations led the authors to conclude that cysteine is an essential growth factor for T. brucei and cannot be replaced by other reducing agents, including D-CYS- teine or cystine.

    Trypanosoma spp. have been found to generate H202 at fairly high rates and to possess high H202 l e ~ e l s . 4 ~ ~ ~ They have long been thought to lack, or to be extremely deficient in, the enzyme systems necessary for the removal of H,O,. Epimastigotes of T. cruzi have been shown to have a deficient capacity to metabolize H202. They were found to lack catalase and glutathione per- oxidase (H20, dependent), but possess SOD44 (Fe-SOD45), GSH, NADPH-dependent glu- tathione reductase, NADPH oxidase, and a very low activity of ascorbate peroxidase with uncertain function.@ It was subsequently shown that glutathione disulfide reduction in trypanosomatids is dependent upon a unique, low-molecular-weight, thiol-contain-

    299

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.

  • ing cofactor (trypanothione), and the NADPH-dependent flavoprotein disulfide reductase (trypanothione reductase) main- tains this metabolite in the dithiol form [T(SH),] within the ell.^,^^ Penketh and Klein4* found that the bloodstream forms of T. b. brucei were not deficient in their ability to metabolize H202, although they were also found to lack catalase, glutathione peroxi- dase, and ascorbate peroxidase. They were found to metabolize H202 at a significant rate (2.6 nmol/mid108 cells), by a mecha- nism different from the host. The mecha- nism involved NADPH and the newly dis- covered cofactor T(SH),. Penketh and Klein48 observed activities of glutathione reductase and NADPH-dependent trypanothione reduc- tase, but a very low activity of trypanothione peroxidase. However, they also found sub- stantial activities of enzymes of the pentose phosphate pathway (e.g., glucose-6-phos- phate dehydrogenase, G-6-PDH), which is a prime candidate for the source of NADPH. The authors therefore suggested that NADPH-dependent H202 consumption (re- duction of H,02 by NADPH) is the major mechanism involved. Henderson et al.49 iden- tified a trypanothione-dependent peroxidase activity in T. brucei and Crithidia fasciculata and considered that this activity represented the sole mechanism for the removal of H,02 in T. brucei. They also expected similar ac- tivities to be present in other species of Try- panosoma and Leishmania that possess the flavoenzyme trypanothione reductase. Penketh et al.50 measured the ability of 12 trypanosomatids to metabolize H202. They confirmed that typical catalase and peroxi- dase hemoproteins were not important in H202 metabolism, and concluded that the novel process responsible for H,O, metabo- lism in T. brucei (NADPH-dependent H20, con~umption~~) was common to many, if not all, trypanosomatids.

    Carnieri et aL5* confirmed that T. cruzi had lirdted ability to detoxify H202; differ-

    ent stages of T. cmzi were found to metabo- lize low concentrations of H202. The epimastigotes of T. cruzi were found to lack trypanothione peroxidase activity but pos- sess NADPH oxidase and NADPH peroxi- dase activities. The NADPH peroxidase ac- tivity was very low, compared with the true peroxidase activities present in mammalian cells, and was probably due to a nonenzy- matic reaction(s) with endogenous T(SH), and/or other thiols. The authors considered that, taking into account the high intracellu- lar concentration of thiols measured, this activity probably accounted for the rates of H20, metabolism detected in intact T. cruzi. Recently, Clark et al.', reported the activity of dehydroascorbate reductase (g1utathione:dehydroascorbate oxidoreduc- tase) in the epimastigotes and trypomastigotes of T. cruzi. The enzyme was known to cata- lyze a glutathione-dependent reduction of dehydroascorbic acid in plant and animal tissues, but its activity had not been reported in protozoa. They also evaluated the vitamin C content and found that the total vitamin C content (ascorbic acid and dehydroascorbic acid) per milligram of protein was I .6 to 3.6 times higher in trypomastigotes than in epimastigotes. Moreover, trypomastigotes also had a predominance of the reduced form (ascorbic acid, 95%), in contrast to the val- ues found in epimastigotes (24 to 47%). This might be related to the higher resistance/ tolerance to in vitro H20, exposure of trypomastigotes than that of epima~tigotes.~~ The presence of dehydroascorbate reductase activity, along with the ascorbate peroxidase activity,44 suggests that an enzymatic vita- min C redox cycle may also be involved in the scavenging reaction in T. cruzi.

    V. LEISHMANIA

    The promastigotes of L. donovani and L. tropica are highly susceptible to H202.54.55

    300

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.

  • H302 at a concentration of lo4 M was found to inhibit the multiplication of L. donovani promastigotes by 94%, and [3H]thymidine and [3H]uracil incorporation by 95 and 97%, respectively. Continuous generation of H,O, by glucose-glucose oxidase also had a pro- found suppressive effect on promastigote proliferation. However, addition of catalase completely abrogated the growth-inhibitory effect.55 The promastigotes were found to be deficient in catalase and glutathione peroxi- dase, but possess SOD.54 They readily trig- ger the macrophage oxidative burst, includ- ing the release of H,O,, and 80 to 95% are promptly killed within the cytoplasm of nor- mal resident cells. However, macrophages inherently deficient in the production of oxygen intermediates (577468 cells) exert no appreciable promastigocidal activity.56 Exposure to concanavalin A-stimulated lym- phokines effectively enhanced the oxidative response of 577468 cells and, similarly, in- duced intracellular antileishmanial activity. Inhibiting macrophage H,O, production con- sistently decreased the killing of Leishmania by lymphokine-treated 577468 cells.56

    Murray et al.57 found that, although ca- pable of killing the promastigotes of L. donovani, normal mouse peritoneal mac- rophages exerted no activity against amastigotes. The amastigotes also survived equally well within the in vivo-activated macrophages. However, they could be eradi- cated by in vitro (1ymphokine)-activated cells. The authors concluded that the suscep- tibility, as well as acquired resistance to L. donovani infection, correlates closely with the host capacity to generate macrophage- activating lymphokines. In the accompany- ing report, Murray5* demonstrated that, com- pared with its promastigote predecessor, L. donovani amastigotes contained threefold more catalase and 14-fold more glutathione peroxidase, and were four times more resis- tant to enzymatically generated H,O,. The intracellular fate of amastigotes was found

    to correlate with two particular macrophage properties: (1) the ability to respond to para- site ingestion with oxidative burst activity and (2) the capacity to generate 0; and H,O,. Macrophages capable of simultaneously dis- playing high levels of both the activities (lym- phokine-activated cells) killed 90% of amastigotes, whereas those demonstrating neither (577468 cells) permitted amastigotes to replicate. Normal resident macrophages and in vivo-activated cells demonstrated ei- ther of the two activities (but not both) and failed to kill amastigotes. Catalase abolished the killing of amastigotes by lymphokine- activated cells, suggesting a primary leishmanicidal role for H202. Extending these observations to human mononuclear phago- cytes and examining the interaction of L. donovani with monocytes, Murray and Ca~-tell i~~ demonstrated that the amastigotes were more resistant than promastigotes to killing by mononuclear phagocytes, and that monocytes were considerably more active against both the forms than the monocyte- derived macrophages. The latter finding ap- peared to reflect the rapid and parallel de- cline in the monocytes oxygen-dependent and -independent antileishmanial mecha- nisms during (their) in vitro cultivation. The activity of both mechanisms, however, could be effectively restored by lymphokine acti- vation.

    In order to determine whether L. dono- vani survives within macrophages by inter- fering with the phagocytic oxidative burst or later oxidative responses, Pearson et aL60 examined the interaction of the promastigote and amastigote stages of the parasite with human monocyte-derived macrophages. They concluded that (1) L. donovani does not survive in macrophages by interfering with the phagocytic oxidative burst, (2) does not produce a continuous oxidative activity, and (3) only transiently alters the subsequent macrophage oxidative responses. In a subse- quent study, Pearson et al.61 found that the

    301

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.

  • survival of L. donovani in human monocytes was dependent on the parasite stage; when promastigotes were ingested, they elicited an oxidative burst, and the majority were killed by oxidative microbicidal mechanisms, whereas amastigotes were ingested and sur- vived to parasitize human monocytes suc- cessfully, despite eliciting a phagocytic oxi- dative burst. In a phagocyte-free system, amastigotes were found to be sevenfold more resistant than were promastigotes to the le- thal effects of H,02. Channon et al.62 dem- onstrated dramatic differences in the bind- ing of, and respiratory burst activity elicited by, promastigotes and amastigotes of L. donovani in murine resident peritoneal macrophages. They explained that, while amastigotes may possess an azide-sensitive mechanism (Fe-SOD, or some other mecha- nism) that either competes for 0; produced or causes localized inactivation of respira- tory burst activity, this cannot account for the full magnitude of the difference between the two forms of the parasite. Transforma- tion and competitive binding studies sug- gested that the more likely explanation lies in both qualitative and quantitative differ- ences in the distribution of surface ligands involved in binding the parasite to the mac- rophage membrane; the well-characterized mannose/fucose receptor may be important in the binding of, and respiratory burst activ- ity induced by, promastigotes but not amastigotes6, Channon and BlackwelP3 de- termined the fate of H202 during its reaction with promastigotes or amastigotes of L. donovani, and also examined the sensi- tivities of both forms of the parasite to re- agent or glucose oxidase-generated H202 in a phagocyte-free system. The amastigotes could remove greater amounts of H20, than the promastigotes. It was also observed that, while promastigotes were equally sensitive to either form of H,Oz stress, amastigotes were more resistant to single, larger amounts of reagent H202 than to equivalent amounts

    of H202 generated over a 1-h period. These results suggested that the differential re- sponses of promastigotes and amastigotes to different forms of H202 stress may depend upon different H20,-scavenging mechanisms. In the accompanying paper, Channon and Blackwell@ detected catalase in amastigotes but not in promastigotes. Glutathione per- oxidase activity was undetectable in either form of the parasite. The total thiol sink (GSH and protein thiols) was greater in promastigotes, but the ability to regenerate GSH via glutathione reductase was equiva- lent for both forms. Less temperature-de- pendent, nonenzymatic mechanisms (e.g., an unsaturated lipid sink) also appeared to con- tribute to the removal of H202 by both forms. The authors, however, supported the idea that the differences in H202 sensitivity be- tween the two forms of the parasite relates to the activity of the direct H,O,-scavenging enzyme, catalase, which appears to operate more efficiently against a bolus of reagent

    The trypanothione system appears to be a central feature of antioxidant defense in trypanosomatids. As discussed earlier, pro- tection against H202 has been proposed to involve a trypanothione-dependent enzymatic reac t i~n .~*-~ ' Phenotypes of recombinant L donovani and T. cruzi have been produced that overexpress trypanothione reductase in response to agents that are thought to induce oxidative stress.65 However, the growth of transformed and control cells was equally sensitive to inhibition by these agents. Likewise, growth of both cell types was equally susceptible to inhibition by H202, and control and transformed L. donovani promastigotes metabolized H202 at compa- rable rates. These experiments suggest that either the reaction normally catalyzed by trypanothione peroxidase is not rate limiting in the clearance of H20, or it is inoperative. Given this, the possibility that a try- panothione-independent system operates for

    H202-

    302

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.

  • the utilization of H,O, should be considered and warrants further investigation.

    The manner in which Leishmania evades intracellular killing is still not completely understood. Indeed, macrophages stimulated with E. coli lipopolysaccharide do secrete large amounts of oxygen metabolites, but they fail to destroy the parasites.& It was thought that Leishmania spp. might interfere with the oxidative metabolism of their host macrophages, and, therefore, the effect of the presence of these intracellular parasites on the respiratory burst of lymphokine-acti- vated and lipopolysaccharide-stimulated mouse macrophages was determined.67 An impairment of the oxidative metabolism of infected vs. noninfected cells was shown; however, the mechanisms of this impairment remained unclear. The parasite has been found to utilize specific receptors (mannose- fucose receptor and the type-three comple- ment receptor CR3) for the attachment and ingestion by r n a c r ~ p h a g e s . ~ ~ - ~ ~ However, the CR3 receptor has been shown to promote phagocytosis but not the generation or re- lease of toxic oxygen intermediates from human phagocy te~ .~~ There is also evidence that the lipophosphoglycan of L. donovani inhibits macrophage protein kinase C activ- ity, an important enzyme that is believed to be responsible for initiating the oxidative b ~ r s t . ~ l - ~ ~ The depletion of protein kinase C rendered macrophages more permissive for the proliferation of L. donovani, suggesting that the inhibition of protein kinase C-de- pendent events contributes to the survival of this parasite within its host cell. The lipophosphoglycan of L. donovani has also been shown to scavenge potentially cyto- . cidal oxygen metabolites in ~ i t i - 0 . ~ ~ These studies suggest that the lipophosphoglycan of Leishmania also contributes in providing protection against oxidative cytocidal mecha- nisms of macrophages. Zarley et al.75 inves- tigated the mechanism of H,O, toxicity for L. d. chagasi promastigotes, and factors re-

    sponsible for their (relative) H,O, resistance. They found that there were at least two dis- tinct mechanisms for the observed H,O, re- sistance in different culture forms of promastigotes. First, as promastigotes grow from the logarithmic to the stationary phase in liquid culture medium, they undergo an increase in their virulence for a murine host as well as enhanced resistance to H,02-me- diated toxicity. Second, exposure of promastigotes to heat shock also caused them to become more resistant to H,O, toxicity, but through an alternate mechanism. Thus, there are both developmental and inducible factors governing H 2 0 2 resistance in promastigotes, and these likely activate dis- tinct resistance mechanisms. Wilson et al.76 have shown that incubation in sublethal con- centrations of H,O, or the redox-cycling agent menadione caused a stress response in promastigotes of L. chagasi. This oxidant- induced response was associated with an increase in the amount of heat-shock protein hsp70. Induction of a stress response by exposure of promastigotes either to heat shock or to sublethal oxidants (H,02 or me- nadione) caused them to become more resis- tant to H20, toxicity, and also more virulent to the murine host. Thus, it may be that the adverse environmental changes encountered naturally during the life-cycle of Leishma- nia species (i.e., heat and oxidant) lead to an increase in resistance to the toxic effects of oxidant exposure, which, in turn, may be critical for the survival of parasite in a mam- malian host.

    VI. PLASMODIUM

    Oxidative stress has been defined as any disturbance of the cellular prooxidanthti- oxidant balance in favor of pro oxidant^.^^ Susceptibility to oxidative stress is a well- established feature of malarial parasites.78 The sensitivity of the parasite to oxygen is

    303

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.

  • demonstrated by the fact that Plasmodium spp. can be successfully cultured only in atmospheres of low p02.79 H,02 treatment tends to reduce the parasitemia both in vitro and in vivo.80,81 Erythrocytes containing Plasmodium parasites trigger production of reactive oxygen species (ROS) by pha- gocytes in vitro. The most direct evidence for the killing of parasites by ROS derived from phagocytes has been with P. yoeliiE2 and P. f a l c ipa r~m;~~ detoxification of ROS was shown to prevent the lethal effects of activated phagocytes. Golenser et al.84 also investigated the role of ROS generated by polymorphonuclear leukocytes in the host response against P. falciparum malaria, and concluded that increased oxidative stress induced by polymorphonuclear leukocytes did interfere with the growth of P. falci- parum. Finally, the fact that oxidative drugs destroy malarial parasites in v i m and in vivo indicates that Plasmodium spp. are suscep- tible to oxidative damage.85

    Plasmodium spp. have mitochondria and utilize oxygen.8c88 Therefore, ROS-generat- ing processes may be present in the parasite, such as the one-electron reduction of oxygen by the electron transport chain of the mito- chondrion. Deslauriers et al.89 investigated the ability of normal and P. berghei-parasit- ized erythrocytes to reduce exogenous free radicals in the form of stable nitroxides. Parasitized red cells were found to reduce the lipid-soluble nitroxide probe at a rate that increased with the level of parasitemia. Inhibitors of electron transport increased the rate of reduction. The authors proposed that the reduction occurred via an electron trans- port chain in the parasite. Production of H202 has been shown in P. berghei,90 but it seems that comparable experiments on other Plas- modium spp. have not been carried out. Hunt and Stocker7 found increased 0,- produc- tion in P. vinckei-parasitized erythrocytes. The autooxidation of lipid that occurs in P. vincket9I and P. falciparum-infected red

    cells92-94 can be taken as (indirect) evidence for oxidant production by the parasite. 1.n an attempt to verify the biochemical origin of oxidative radicals in malaria-infected eryth- rocytes, Atamna and Ginsburgg5 found that ROS are produced in the food vacuole of the parasite during the digestion of host cell cytosol, and the H,02 produced during the digestion reaches the host cell compartment. Har-El et al.% suggested that induction of oxidant stress could be mediated by redox- active hemes that are being supplied in an increasing amount by the maturing parasite as it degrades host hemoglobin. They pro- posed that hemin or hemin-like structures are the appropriate candidates that could catalyze oxidative stress and deregulate the delicate redox balance of the host-parasite system. These s t u d i e ~ ~ ~ ? ~ ~ reestablish the fact that the malarial parasite inflicts an oxida- tive stress on its host cell, demonstrate the biochemical origin of this stress, and sup- port the idea that heme is involved in the susceptibility of Plasmodium to oxidant stress.

    Plasmodium is endowed with its own antioxidant defense mechanisms. Isolated P. berghei parasites were not found to con- tain endogenous SOD. Host SOD was im- ported and concentrated in the parasites ly- sosomes. In addition, isolated P. berghei also lacked c a t a l a ~ e . ~ ~ , ~ ~ Glutathione reductase has been reported in isolated P. berghei.% In other studies,lOOJO1 isolated P. berghei has been found to contain significant amounts of GSH, glutathione reductase, glutathione peroxi- dase, and glutathione-S-transferase. Isolated early intraerythrocytic stages of P. falci- parum contain catalase and SOD but little or no glutathione peroxidase. However, isolated late-intraerythrocytic stages contain slightly less catalase but significantly more glu- tathione peroxidase and SOD. P. falciparum, like P. berghei,97 probably acquires most of its SOD from its host because parasite-asso- ciated SOD is predominantly cyanide-sensi-

    304

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.

  • tive (Cu.Zn-SOD). Unlike P. berghei, how- ever, late stages of P. falciparum contain an additional SOD isozyme that is not cyanide sensitive (Fe- or Mn-SOD), and may repre- sent an endogenous enzyme. Parasites grown in red cells that have been partially depleted of SOD were found to be more sensitive to exogenously generated 03, suggesting some dependence of the parasite on the host SOD.lo2 Ranz and Meshnick103 reported that the endogenous P. falciparum SOD isozyme was most likely Mn-SOD because it was H202 insensitive, and was probably mito- chondrial in origin. Golenser et a1.lW found that this plasmodial SOD existed in various strains of P. falciparum, but the majority of SOD activity found in the parasite was re- lated to the host enzyme. Becuwe et al.Io5 found a cyanide-resistant, H,02-sensitive endogenous Fe-SOD in P. berghei, P. yoelii, and P. vinckei. This endogenous activity rep- resented 25 to 30% of the total SOD activity; the remaining activity may correspond to the internalized RBC Cu.Zn-SOD. The authors suggested that these rodent malarial para- sites may be capable of at least partly resist- ing activated oxygen species using an en- dogenous SOD. Recently, Becuwe et al.06 characterized a cyanide-resistant, H,O,-sen- sitive endogenous Fe-SOD in P. fakiparum that represented 20 to 30% of the total SOD activity found in the crude extract. The re- maining 80% activity may correspond to the RBC SOD, internalized in the digestive sys- tem of the parasite. According to these au- thors, the Fe-SOD activity found in P. falciparum represents the first level of antioxidant defense of the parasite. P. falci- parum also has its own glutathione reduc- tase, and it has been speculated that the para- site can store host glutathione reductase during the course of intraerythrocytic matu- ration.lo7 Recently, the antimalarial activity of a number of glutathione reductase inhibi- tors was established,lo8 and identification and sequence analysis of the glutathione reduc-

    tase gene has been reported.Iw The presence of G-6-PDH in Plasmodium has been a sub- ject of controversy for many years, but it now seems to be convincingly established and partially characterized in P. berghei1I0 and in P. falciparum. Pentose phosphate pathway (or hexose monophosphate shunt, HMS) activity has been measured in intact infected red blood cells.Il2 Recently, Atamna et a l . I l 3 demonstrated considerable HMS ac- tivity in free P. falciparum that was about 8 1 to 82% of the total activity seen in parasite- infected red cells. The HMS activity of these infected red cells increased with parasite maturation, which may reflect the oxidative stress generated by the proliferating para- site.

    Plasmodium spp. synthesize a number of histidine-rich proteins (HRPs) .~~~ His- tidine analogs have been shown to inhibit the growth of P. falciparum in vitro,117J18 suggesting that HRPs play important role(s) in parasite survival. Histidine is a special agent that can intervene in free-radical reac- tions in a variety of modes, and can act as a protecting agent.lI9 It has been observed that histidine could prevent P. falcipanun destruc- tion in vitro by freshly isolated and activated human polymorphonuclear neutrophils.120 Based on these observations, it is intriguing to suggest that HRPs may serve an antioxi- dant function for the parasite.

    The malarial parasite derives a number of biochemical advantages from its host erythrocytes, particularly hemoglobin as a source of nutrients and a set of red cell en- zymes whose activities can be utilized. From the literature, it seems clear that the parasite produces oxidative radicals that diffuse into the host cell and disturb its redox equilib- rium. Hence, both the hosts and the parasites antioxidant defense systems, must be con- siderably activated to counteract this oxida- tive challenge. The energy metabolism of the parasite-infected red cells and its role(s) in maintaining the redox status have been

    305

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.

  • reviewed by Ginsburg,l2' Hunt and Stocker,77 and Roth,l12 suggesting, in general, an upregulation of certain key antioxidant mechanisms. Recently, evidence has been presented for the increased uptake and me- tabolism of riboflavin,122 GSH, vitamin C and vitamin E ~ o n t e n t s , ~ ~ J ~ * ~ ~ and HMS activity' l3 in Plasmodium-infected erythro- cytes. Thus, the overall ability of the parasite to withstand oxidant stress in its microenvi- ronment is determined by a number of fac- tors, and seems to be related to the exploita- tion of the erythrocyte antioxidant defense enzymes as well as activatiordproduction of its own protective mechanisms.

    VII. CONCLUSIONS AND FUTURE PERSPECTIVES

    Careful analysis of the literature indi- cates the following important characteristics of oxidant-defense systems of parasitic pro- tozoa:

    1.

    2.

    3.

    Catalase is absent in Entamoeba histolytica, Giardia, and Trypanosoma. Other metabolic pathways for H202 detoxification exist in these organisms, some of which are similar to those of certain prokaryotes. Cysteine seems to be an essential growth factor for E. histolytica, Giardia, and T. brucei. Giardia and T. brucei cannot transport cystine and cannot synthesize cysteine de novo. Similar studies, how- ever, have not been carried out for E. histolytica. E. histolytica and Giardia (and tri- chomonads) lack GSH and associated enzymes, but possess high levels of cysteine. Therefore, cysteine-mediated, nonenzymatic reactions seem to func- tion as the detoxification mechanism in these organisms. They have also been stiown to possess alternative pathways

    from those described for aerobic eu- karyotes. The functional significance of these alternative mechanisms (294 34-kDa protein in E. histolytica, and NADH oxidase and NADH peroxidase in Giardia) has been shown in Salmo- nella typhimurium126 and Haemophilus somnus and Streptococcus faecalis. 127 Trypanosomes need cysteine not only for protein biosynthesis but also for GSH and T(SH), formation, which are present in high amounts. Besides trypanothione metabolism, T. cruzi has been shown to possess an ascorbate peroxidase/dehydroascorbate reductase system as a scavenging mechanism. Ascorbate peroxidase has been found to be effective in scavenging H,O, in various species of cyanobacteria and eukaryotic algae.lZ8 Recently, T. foetus and T. vaginalis were also shown to contain ascorbate peroxidase, which may scavenge low concentrations of Hz02.129 Leishmania has been shown to possess certain nonenzymatic, oxidant-scaveng- ing mechanisms. These include, the surface glycolipid scavenger lipophos- phoglycan, and induction of heat-shock proteins in response to heat and oxidant stress. Mycobacterium leprae,74 Salmo- nella typhim~rium'~~ and Escherichia c01i131J32 have already been found to be equipped with such mechanisms. Re- cently, proteoglycans from bovine cor- nea showed a protective effect on lipo- some peroxidation induced by Fez+, supporting the hypothesis that proteo- glycans may represent part of the anti- oxidant mechanisms of organisms.133 Plasmodium utilizes a variety of host enzymes for its own protection, al- though the parasite also has its own defense system. Although a different organism, an analogous situation may exist in Entamoeba histolytica, which

    4.

    5 .

    6.

    306

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.

  • engulfs bacteria during its commensal phase and RBCs during its invasive phase. It has been hypothesized that the engulfed bacteria and erythrocytes, ei- ther intact and/or through their (anti- oxidant) components, detoxify the prod- ucts of oxygen reduction to protect the amoebae during i n v a ~ i o n . l ~ ~ ~ ~ How- ever, a possible mechanism of how these components might function within the organism has not been proposed so far.

    Beside Leishmania, other species (e.g., E. histolytica, Giardia, Trypanosoma, and Plasmodium) also contain lipophosphoglycan or lipophosphoglycan-like m o l e ~ u l e s . ~ ~ ~ J ~ ~ Similarly, E. histolytica, 139 Giardia, I4O try- pa no some^,^^^ and Plasmodi~rn~*-~~ have also been shown to contain heat-shock pro- teins. However, the functional significance of heat-shock proteins and lipophos- phoglycan as antioxidant mechanisms in these organisms has not been explored. It would be interesting to determine whether, as in Leishmania and certain prokaryotes, these molecules provide a (secondary) de- fense against oxidative stress.

    Thus, parasitic protozoa have a diverse arsenal of antioxidant defenses, and many of these are shared with the prokaryotes. Re- cent developments in cloning specific genes, and the isolation of regulatory mutants have helped further our understanding of the regu- lation of these defense mechanisms in prokaryotes. Analogous studies among the parasitic protozoa hold the promise of being similarly fruitful.

    ACKNOWLEDGMENTS

    My sincere thanks are due to Dr. Eric Pearlman, Assistant Professor, Division of Geographic Medicine and Department of Ophthalmology, for his constant support and encouragement, and his valuable coopera-

    tion and help in various ways in completing this review. I also want to recognize the contribution of Dr. Wes Kroeze of the De- partment of Biochemistry, who has read the manuscript and contributed valuable sugges- tions and criticisms. Without such help, this review would indeed be poorer.

    REFERENCES

    1.

    2.

    3.

    4.

    5.

    6.

    7.

    8.

    9.

    Hassan, H. M., Microbial superoxide dismutases, Adv. Genet., 26, 65, 1989.

    James, E. R., Superoxide dismutase, Parasitol. Today, 10, 481, 1994.

    Kroll, J. S., Langford, P. R., Wilks, K. E., and Keil, A. D., Bacterial (Cu.Zn)-superox- ide dismutase: phylogenetically distinct from the eukaryotic enzyme, and not so rare after all!, Microbiology, 141, 2271, 1995.

    Benov, L. and Fridovich, I., Functional sig- nificance of the Cu.Zn-SOD in Escherichia coli, Arch. Biochem. Biophys., 327,249, 1996.

    Diamond, L. S., Techniques of axenic culti- vation of Entamoeba histolytica Schaudinn, 1903 and E. histolytica-like amoebae, J. Parasitol., 54, 715, 1968.

    Diamond, L. S., Harlow, D. R., and Cunnick, C. C., A new medium for the axenic cultivation of Entamoeba histolytica and other Entamoebae, Trans. R. SOC. Trop. Med. Hyg., 72, 431, 1978.

    Singh, B. N., Das, S. R., and Dutta, G. P., Importance of 0-R potential in initiating cul- tures of axenically grown Entamoeba histolytica from small inocula, Curr. Sci., 42, 227, 1973.

    Singh, B. N., Dutta, G. P., and Das, S. R., Effect of 0 - R potential on the growth and multiplication of axenic Entamoeba histolytica, Curr. Sci., 43, 71, 1974.

    Gillin, F. D. and Diamond, L.S., Entam- oeba histolytica and Giardia lamblia: growth responses to reducing agents, Exp. Parasitol., 51, 382, 1981.

    307

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.

  • 10. Reeves, R. E., Metabolism of Entamoeba histolytica Schaudinn, 1903, Adv. Parasitol., 23, 105, 1984.

    21. Mehlotra, R. K. and Shukla, 0. P., Reduc- ing agents and Entamoeba histolytica, Parasitol. Today, 4, 82, 1988.

    22. Reed, S. L., Flores, B. M., Batzer, M. A., Stein, M. A., Stroeher, V. L., Carlton, J. E., Diedrich, D. L., and Torian, B. E., Molecu- lar and cellular characterization of the 29- kflodalton peripheral membrane protein of Entamoeba histolytica: differentiation be- tween pathogenic and non-pathogenic isolates, Infect. Immun., 60, 542, 1992.

    23. Bruchhaus, I. and Tannich, E., Analysis of the genomic sequence encoding the 29-kDa cy steine-rich protein of Entamoeba histolytica, Trop. Med. Parasitol., 44, 116, 1993.

    11. Mehlotra, R. K., Shukla, 0. P., and Singh, B. N., pH as the critical factor in cultivation of Entamoeba histolytica, Ind. J. Parasitol., 10, 135, 1986.

    12. Weinbach, E. C. and Diamond, L. S., Enta- moeba histolytica. I. Aerobic metabolism, Exp. Parasitol., 35, 232, 1974.

    13. Band, R. N. and Cimto, H., Growth re- sponses of axenic Entamoeba histolytica to hydrogen, carbon dioxide and oxygen, J. Protozool., 26, 282, 1979.

    14. Sykes, D. E. and Band, R. N., Superoxide dismutase activity of Acanthamoeba and two anaerobic Entamoeba species, J. Cell Biol., 75, 86a, 1977.

    15. Fahey, R. C., Newton, G. L., Amck, B., Overdank-Bogart, T., and Aley, S. B., En- tamoeba histolytica: a eukaryote without glu- tathione metabolism, Science, 224,70, 1984.

    16. Tannich, E., Bruchhaus, I., Walter, R D., and Horstmann, R. D., Pathogenic and non- pathogenic Entamoeba histolytica: identifi- cation and molecular cloning of an iron-con- taining superoxide dismutase, Mol. Biochem. Parasitol., 49, 61, 1991.

    17. Murray, H. W., Aley, S. B., and Scott, W. A., Susceptibility of Entamoeba histolytica to oxygen intermediates, Mol. Biochem. Parasitol., 3, 381, 1981.

    18. Ghadirian, E., Somerfield, S. D., and Kongshavn, P. A. L., Susceptibility of Enta- moeba histolytica to oxidants, Infect. Immun., 51, 263, 1986.

    19. Hungate, R. E., A roll tube method for cul- tivation of strict anaerobes, in Methods in Microbiology, 3B, Noms, J. R. and Ribbons, D. W., Eds., Academic Press, New York, 1969, 117.

    24. Bruchhaus, I. and Tannich, E., Identifica- tion of an Entamoeba histolytica gene encod- ing a protein homologous to prokaryotic disulphide oxidoreductases, Mol. Biochem. Parasitol., 70, 187, 1995.

    25. Bruchhaus, I. and Tannich, E., Induction of the iron-containing superoxide dismutase in Entamoeba histolytica by a superoxide an- ion-generating system or by iron chelation, Mol. Biochem. Parasitol., 67, 281, 1994.

    26. Visvesvara, G. S., Axenic growth of Giardia lamblia in Diamonds TP-S- 1 medium, Trans. R. SOC. Trop. Med. Hyg., 74, 213, 1980.

    27. Keister, D. B., Axenic culture of Giardia lamblia in TYI-S-33 medium supplemented with bile, Trans. R. SOC. Trop. Med. Hyg., 77, 487, 1983.

    28. Weinbach, E. C., Claggett, C. E., Keister, D. B., Diamond, L. S., and Kon, H., Respi- ratory metabolism of Giardia lamblia. J. Parasitol., 66, 347, 1980.

    29. Paget, T. A., Kelly, M. L., Jarroll, E. L., Lindmark, D. G., and Lloyd, D., The ef- fects of oxygen on fermentation in Giardia lamblia, Mol. Biochem. Parasitol., 57, 65, 1993.

    30. Brown, D. M., Upcroft, J. A., and Upcroft, P., Cysteine is the major low-molecular weight thiol in Giardia duodenalis, Mol. Biochem. Parasitol., 61, 155, 1993.

    3 1. Brown, D. M., Upcroft, J. A., and Upcroft, P., Free radical detoxification in Giardia

    20. Gillin, F. D. and Diamond, L. S., Entam- oeba histolytica and Giardia lamblia: effects of cysteine and oxygen tension on trophozo- ite attachment and survival in culture media, Exp. Parasitol., 52, 9, 1981.

    308

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.

  • duodenalis, Mol. Biochem. Parasitol., 72,47, 1995.

    32. Gillin, F. D., Reiner, D. S., Levy, R. B., and Henkart, P. A., Thiol groups on the surface of anaerobic parasitic protozoa, Mol. Biochem. Parasitol., 13, 1, 1984.

    33. Lujan, H. D. and Nash, T. E., The uptake and metabolism of cysteine by Giardia lamblia trophozoites, J. Euk Microbiol., 4 1, 169, 1994.

    34. Fairlamb, A. H., Novel biochemical path- ways in parasitic protozoa, Parasitology, 99. S93, 1989.

    35. Lo, H.4. and Reeves, R. E., Purification and properties of NADPH:flavin oxidoreduc- tase from Entamoeba histolytica, Mol. Biochem. Parasitol., 2, 23, 1980.

    36. Linstead, D. J. and Bradley, S., The purifi- cation and properties of two soluble reduced nicotinamide:acceptor oxidoreductases from Trichomonas vaginalis, Mol. Biochem. Parasitol., 27, 125, 1988.

    42. Meshnick, S. R., Chang, K.-P., and Cerami, A., Heme lysis of the bloodstream forms of Ttypanosoma brucei, Biochem. Pharmacol., 26, 1923, 1977.

    43. Meshnick, S. R., Blobstein, S. H., Grady, R. W., and Cerami, A., An approach to the development of new drugs for African trypa- nosomiasis, J. Exp. Med., 148, 569, 1978.

    44. Boveris, A., Sies, H., Martino, E. E., Docampo, R., Turrens, J. F., and Stoppani, A. 0. M., Deficient metabolic utilization of hydrogen peroxide in Trypanosoma cruzi, Biochem. J.. 188, 643, 1980.

    45. Temperton, N. J., Wilkinson, S. R., and Kelly, L. M., Cloning of an Fe-superoxide dismutase gene homologue from Trypano- soma cruzi, Mol. Biochem. Parasitol., 76,339, 1996.

    46. Fairlamb, A. and Cerami, A., Identification of a novel, thiol-containing co-factor essen- tial for glutathione reductase enzyme activity in trypanosomatids, Mol, Biochem. Parasitol., 14, 187, 1985.

    37 Ba1tz9 T.9 Baltz D.7 Giroud9 G9 and Crockett, J., Cultivation in a semi-defined medium of animal infective forms of Trypa- nosoma brucei, T. equiperdum, T. evansi, T. rhodesiense and T. gambiense, EMBO J., 4, 1273, 1985.

    47. Fairlamb, A., Blackburn, P., Ulrich, P., Chait, B., and Cerami, A., Trypanothione: a novel bis (glutathionyl) spermidine co-factor for glutathione reductase in uypanosomatids, Science, 22, 1485, 1985.

    48. Penketh, P. G. and Klein, R. A., Hydrogen peroxide metabolism in Trypanosoma brucei, Mol. Biochem. Parasitol., 20, 111, 1986.

    38. Duszenko, M., Ferguson, M. A. J., Lamont, G. S., Rifkin, M. R., and Cross, G. A. M., Cysteine eliminates the feeder cell require- ment for cultivation of Trypanosoma brucei bloodstream forms in vitro, J . Exp. Med., 162, 1256, 1985.

    49. Henderson, G. B., Fairlamb, A. H., and Cerami, A., Trypanothione dependent per- oxide metabolism in Crithidia fasciculata

    39. Hamm, B., Schindler, A., Mecke, D., and Duszenko, M., Differentiation of Trypano-

    and Trypanosoma brucei, Mol. Biochem. Parasitol., 24, 39, 1987. _ _

    soma brucei bloodstream trypomastigotes from long slender to short stumpy-like forms in axenic culture, Mol. Biochem. Parasitol., 40, 13, 1990. 221,427, 1987.

    40. Duszenko, M., Muhlstadt, K., and Broder, A., Cysteine is an essential growth factor for Trypanosoma brucei bloodstream forms, Mol. Biochem. Parasitol., 50, 269, 1992.

    41. Boveris, A. and Stoppani, A. 0. M., Hydro- gen peroxide generation in Trypanosoma cruzi, Experientia, 33, 1306, 1977.

    50. Penketh, P. G., Kennedy, W. P. K., Patton, C. L., and Sartorelli, A. C., Trypanosomatid hydrogen peroxide metabolism, FEBS Lett.,

    51. Carnieri, E. G. S., Moreno, S. N. J., and Docampo, R., Trypanothione-dependent per- oxide metabolism in Ttypanosoma cruzi dif- ferent stages, Mol. Biochem. Parasitol., 61, 79, 1993.

    52. Clark, D., Albrecht, M., and Arevalo, J., Ascorbate variations and dehydroascorbate

    309

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.

  • reductase activity in Trypanosoma cruzi epimastigotes and trypomastigotes, Mol. Biochem. Parasitol., 66, 143, 1994.

    62. Channon, J. Y., Roberts, M. B., and Blackwell, J. M., A study of the differential respiratory burst activity elicited by pro-

    53. Nathan, C., Nogueira, N., Juangbhanich, C., Ellis, J., and Cohn, Z., Activation of macrophages in vivo and in vitro: correlation between hydrogen peroxide release and kill- ing of T. cmzi, J. Exp. Med., 149,1056, 1979.

    54. Murray, H. W., Susceptibility of Leishma- nia to oxygen intermediates and killing by normal macrophages, J. Exp. Med., 153,1302, 1981.

    55. Reiner, N. E. and Kazura, J. W., Oxidant- mediated damage of Leishmania donovani promastigotes, Infect. Imrnun., 36,1023,1982.

    56. Murray, H. W., Interaction of Leishmania with a macrophage cell-line: correlation be- tween intracellular killing and the generation of oxygen intermediates, J. Exp. Med., 153, 1690, 1981.

    57. Murray, H. W., Masur, H., and Keithly, J. S., Cell-mediated immune response in ex- perimental visceral leishmaniasis. I. Correla- tion between resistance to Leishmania donovani and lymphokine-generating capac- ity, J. Immunol., 129, 344, 1982.

    58. Murray, H. W., Cell-mediated immune re- sponse in experimental visceral leishmania- sis. 11. Oxygen-dependent killing of intracel- lular Leishmania donovani amastigotes, J. Immunol., 129, 351, 1982.

    m&tigotes and amastigotes of Leishmania donovani in murine resident peritoneal mac- rophages, Immunology, 53, 345, 1984.

    63. Channon, J. Y. and Blackwell, J. M., A siudy of the sensitivity of Leishmania donovani promastigotes and amastigotes to hydrogen peroxide. I. Differences in sensitiv- ity correlate with parasite-mediated removal of hydrogen peroxide, Parasitology, 9 1, 197, 1985.

    64. Channon, J. Y. and Blackwell, J. M., A study of the sensitivity of Leishmania donovani promastigotes and amastigotes to hydrogen peroxide. II. Possible mechanisms involved in protective H,O, scavenging, Para- sitology, 91, 207, 1985.

    65. Kelly, J. M., Taylor, M. C., Smith, K., Hunter, K. J., and Fairlamb, A. H., Pheno- type of recombinant Leishmania donovani and Trypanosoma cruzi which over-express trypanothione reductase: sensitivity towards agents that are thought to induce oxidative stress, Eur. J. Biochem., 218, 29, 1993.

    66. Buchmiiller-Rouiller, Y. and Mauel, J., Correlation between enhanced oxidative me- tabolism and leishmanicidal activity in acti- vated macrophages from healer and non-healer mouse strains, J. Immunol., 136, 3884, 1986.

    67. Buchmiiller-Rouiller, Y. and Mauel, J., Impairment of the oxidative metabolism of mouse peritoneal macrophages by intracellu- lar Leishmania spp., Infect. Immun., 5 5 , 587, 1987.

    68. Blackwell, J. M., Ezekowitz, R. A. B., Rob- erts, M. B., Channon, J. Y., Sim, R. B., and

    59. Murray, H. W. and Cartelli, D. M., Killing of intracellular Leishmania donovani by hu- man mononuclear phagocytes: evidence for oxygen-dependent and -independent leish- manicidal activity, J. Clin. Invest., 72, 32, 1983.

    60. Pearson, R. D., Harcus, J. L., Symes, P. H., Romito, R., and Donowitz, G. R., Failure of the phagocytic oxidative response to protect human monocyte-derived macrophages from infection by Leishmania donovani, J. Immunol., 129, 1282, 1982.

    61. Pearson, R. D., Harcus, J. L., Roberts, D., and Donowitz, G. R., Differential survival of Leishmania donovani amastigotes in hu- man monocytes, J. Immunol., 131, 1994, 1983.

    Gordan, S., Macrophage complement and lectin-like receptors bind Leishmania in the absence of serum, J. Exp. Med., 162, 324, 1985.

    69. Wilson, M. E. and Pearson, R. D., Roles of CR3 and mannose receptors in the attach- ment and ingestion of LRishmania donovani by human mononuclear phagocytes, Infect. Immun., 56, 363, 1988.

    70. Wright, S. D. and Silverstein, S. C., Recep- tors for C3b and C3bi promote phagocytosis

    31 0

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.

  • but not the release of toxic oxygen from hu- man phagocytes, J. Exp. Med., 158, 2016, 1983.

    71. McNeely, T. B. and Turco, S. J., Inhibition of protein kinase C activity by the Leishma- nia donovani lipophosphoglycan, Biochem. Biophys. Res. Commun., 148, 653, 1987.

    72. Descoteaux, A., Turco, S. J., Sacks, D. L.,

    73.

    74.

    75.

    76.

    77.

    78.

    79.

    80.

    and Matlashewski, G., Leishmania donovani lipophosphoglycan selectively inhibits signal transduction in macrophages, J. Immunol., 146, 2747, 1991.

    Descoteaux, A., Matlashewski, G., and Turco, S. J., Inhibition of macrophage pro- tein kinase C-mediated protein phosphoryla- tion by Leishmania donovani lipophos- phoglycan, J. Immunol., 149, 3008, 1992.

    Chan, J., Fuziwara, T., Brennan, P., McNeil, M., Turco, S. J., Sibille, J.-C., Snapper, M., Aisen, P., and Bloom, B., Microbial glycolipids: possible virulence fac- tors that scavenge oxygen radicals, Proc. Natl. Acad. Sci. U.S.A., 86, 2453, 1989.

    Zarley, J. H., Britigan, B. E., and Wilson, M. E., Hydrogen peroxide-mediated toxicity for Leishmania donovani chagasi pro- mastigotes: role of hydroxy radical and pro- tection by heat shock, J. Clin. Invest., 88, 1511, 1991.

    Wilson, M. E., Andersen, K. A., and Britigan, B. E., Response of Leishmania chagasi promastigotes to oxidant stress, In- fect. Immun., 62, 5133, 1994.

    Hunt, N. H. and Stocker, R., Oxidative stress and the redox status of malaria-infected eryth- rocytes, Blood Cells, 16, 499, 1990.

    Clark, I. A., Chaudhri, G., and Cowden, W. B., Some roles of free radicals in malaria, Free Rad. Biol. Med., 6, 3 15, 1989.

    Scheibel, L. W., Ashton, S. H., and Trager, W., Plasmodium falciparum: microaerophilic requirements in human blood cells, Enp. Parasitol., 47, 410, 1979.

    Clark, I. A., Cowden, W. B., and Butcher, G. A., Free oxygen radical generators as an- timalarial drugs, Lancet, 1, 234, 1983.

    81. Dockrell, H. M. and Playfair, J. H. L., Kill- ing of blood-stage murine malaria parasites by hydrogen peroxide, Infect. Immun., 39, 456, 1983.

    82. Ockenhouse, C. F. and Shear, H. L., Oxida- tive killing of the intraerythrocytic malaria parasite Plasmodium yoelli by activated mac- rophages, J. Immunol., 132, 424, 1984.

    83. Ockenhouse, C. F., Schulman, S., and Shear, H. L., Induction of crisis forms in the human malaria parasite Plasmodium fulciparum by y-interferon-activated, mono- cyte-derived macrophages, J. Immunol., 133, 1601, 1984.

    84. Golenser, J., Kamyl, M., Tsafack, A., Marva, E., Cohen, A., Kitrossky, N., and Chevion, M., Correlation between destruc- tion of malarial parasites by polymorpho- nuclear leucocytes and oxidative stress, Free Rad. Res. Commun., 17, 249, 1992.

    85. Meshnick, S. R., Jefford, C. W., Posner, G. H., Avery, M. A., and Peters, W., Sec- ond-generation antimalarial endoperoxides, Parasitol. Today, 12, 79, 1996.

    86. Deslauriers, R., Geoffrion, Y., Butler, K. W., and Smith, I. C. P., Magnetic reso- nance studies of the pathophysiology of mu- rine malaria, Q. Rev. Biophys., 18, 65, 1985.

    87. Deslauriers, R., Moffatt, D. J., and Smith, I. C. P., Oxygen consumption in Plasmodium berghei-infected murine red cells: a direct spectrophotometric assay in intact erythro- cytes, Biochim. Biophys. Acta, 886,3 19, 1986.

    88. Fry, M. and Beesley, J. E., Mitochondria of mammalian Plasmodium spp., Parasitology, 102, 17, 1991.

    89. Deslauriers, R., Butler, K., and Smith, I. C. P., Oxidant stress in malaria as probed by stable nitroxide radicals in erythrocytes in- fected with Plasmodium berghei: the effects of primaquine and chloroquine, Biochim. Biophys. Acta, 931, 267, 1987.

    90. Etkin, N. L. and Eaton, J. W., Malaria-in- duced erythrocyte oxidant sensitivity, in Eryth- rocyte Structure and Function, Brewer, G . J., Ed., Alan R. Liss, New York, 1975, 219.

    31 1

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.

  • 91. Buffinton, G. D., Hunt, N. H., Cowden, W. B., and Clark, I. A., Detection of short-chain carbonyl products of peroxidation from ma- laria-parasite (Plasmodium vinckei)-infected red blood cells exposed to oxidative stress, Biochem. J.. 249, 63, 1988.

    92. Mohan, K., Ganguly, N. K., Dubey, M. L., and Mahajan, R. C., Oxidative damage of erythrocytes infected with Plasmodium falciparum: an in vifro study, Ann. Hematol., 65, 131, 1992.

    93. Shoes, A. P. F. C., van den Berg, J. J. M., Roelofsen, B., and Op den Kamp, J. A. F., Lipid peroxidation in Plasmodium falcipanun- parasitized human erythrocytes, Arch. Biochem. Biophys., 298, 651, 1992.

    94. Wozencraft, A. O., Damage to malaria-in- fected erythrocytes following exposure to oxidant-generating systems, Parasitology, 92, 559, 1986.

    95. Ginsburg, H. and Atamna, H., Origin of reactive oxygen species in erythrocytes in- fected with Plasmodium falciparum, Mol. Biochem. Parasitol., 61, 231, 1993.

    96. Har-El, R., Marva, E., Chevion, M., and Golenser, J., Is hemin responsible for the susceptibility of Plasmodia to oxidant stress?, Free Rad. Res. Commun., 18, 279, 1993.

    97. Fairfield, A. S., Meshnick, S. R., and Eaton, J. W., Malaria parasites adopt host cell su- peroxide dismutase, Science, 221.764, 1983.

    98. Fairfield, A. S., Eaton, J. W., and Meshnick, S. R., Superoxide dismutase and catalase in the murine malaria, Plasmodium berghei: content and sub-cellular distribution, Arch. Biochem. Biophys., 250, 526, 1986.

    99. Eckman, J. R., Glutathione metabolism in malaria infected erythrocytes, in Malaria and the Red Cell, Eaton, J. W. and Brewer, G. J., Eds., Alan R. Liss, New York, 1984, 1.

    100. Seth, R. K., Saini, A. S., and Jaswal, T. S., Plasmodium berghei: oxidant defense sys- tem, Exp. Parasitol., 60, 414, 1985.

    101. Dubois, V. L., Platel, D. F. N., Pauly, G., and Tribouley-Duret, J., Plasmodium berghei: implication of intracellular glu- tathione and its related enzyme in chloro-

    quine resistance in vivo, Exp. Parasitol., 81, 117, 1995.

    102. Fairfield, A. S., Abosch, A., Ranz, A., Eaton, J. W., and Meshnick, S. R., Oxidant defense enzymes of Plasmodium falciparum, Mol. Biochem Parasitol., 30, 77, 1988.

    103. Ranz, A. and Meshnick, S. R., Plasmodium falcipanun: inhibitor sensitivity of the endo- genous superoxide dismutase, Exp. Parasitol., 69, 125, 1989.

    104. Golenser, J., Marva, E., and Chevion, M., The survival of Plasmodium under oxidant stress, Parasitol. Today, 7, 142, 1991.

    105. Becuwe, P., Solmianny, C., Camus, D., and Dive, D., Presence of an endogenous super- oxide dismutase activity in three rodent ma- laria species, Parasitol. Res., 79, 349, 1993.

    106. Becuwe, P., Gratepanche, S., Fourmaux, M.-N., Beeumen, J. V., Samyn, B., Mercereau-Puijalon, O., Touzel, J. P., Slomianny, C., Camus, D., and Dive, D., Characterization of iron-dependent endog- enous superoxide dismutase of Plasmodium falcipanun, Mol. Biochem Parasitol., 76, 125, 1996.

    107. Hempelmann, E., Schirmer, R. H., Fritsch, G., Hundt, E., and Groschel-Stewart, U., Studies on glutathione reductase and meth- emoglobin from human erythrocytes parasit- ized with Plasmodium falciparum, Mol. Biochem. Parasitol., 23, 19, 1987.

    108. Schirmer, R. H.,.Miiller, J. G., and Krauth- Siegel, R L., Disulfide-reductase inhibitors as chemotherapeutic agents: the design of drugs for trypanosomiasis and malaria, Angew. Chem. Int. Ed. Engl., 34, 141, 1995.

    109. Miiller, S., Becker, K., Bergmann, B., Schirmer, R H., and Walter, 2. D., Plas- modium falciparum glutathione reductase exhibits sequence similarities with the human host enzyme in the core structure but differs at the ligand-binding sites, Mol. Biochem. Parasitol., 74, 11, 1995.

    110. Buckwitz, D., Jacobash, G., Kuckelkorn, U., Plonka, A., and Gerth, C., Glucose-6- phosphate dehydrogenase from Plasmodium berghei: kinetic and electrophoretic charac- terization, Exp. Parasitol., 70, 264, 1990.

    312

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.

  • 1 1 I . Kurdi-Haider, B. and Luzzatto, L., Expres- sion and characterization of glucose-6- phosphate dehydrogenase of Plasmodium falciparum, Mol. Biochem. Parasitol., 41,83, 1990.

    1 12. Roth, E., Jr., Plasmodium falciparum carbo- hydrate metabolism: a connection between host cell and parasite, Blood Cells, 16, 453, 1990.

    1 13. Atamna, H., Pascarmona, G., and Ginsburg, H., Hexose-monophosphate shunt activity in intact Plasmodium falciparum-infected eryth- rocytes and in free parasites, Mol. Biochem. Parasitol., 67, 79, 1994.

    114. Ravetch, J. V., Feder, R., Pavlovec, A., and Blobel, G., Primary structure and genomic organization of the histidine-rich protein of the malaria parasite Plasmodium lophurue, Nature (London), 312, 616, 1984.

    115. Howard, R. J., Uni, S., Aikawa, M., Aley, S. B., Leech, J. H., Lew, A. M., Wellems, T. E., Rener, J., and Taylor, D. W., Secre- tion of a malarial histidine-rich protein (PfHRP 11) from Plasmodium falciparum-infected erythrocytes, J. Cell Bid., 103, 1269, 1986.

    116. Kilejian, A., Yang, Y.-F., Cochrane, A. H., and Rashid, M. A., Homologous sequences in Plasmodium cynomolgi and the gene of the histidine-rich knob protein of Plasmodium falciparum, Mol. Biochem. Parasitol., 38,29 1, 1990.

    117. Howard, R. J., Andrutis, A. T., Leech, J. H., Ellis, W. Y., Cohen, L. A., and Kirk, K. L.,

    120. Nnalue, N. A. and Friedman, M. J., Evi- dence for a neutrophil-mediated protective response in malaria, Parasite Immunol., 10, 47, 1988.

    12 1. Ginsburg, H., Some reflections concerning host erythrocyte-malarial parasite interrela- tionships, Blood Cells, 16, 225, 1990.

    122. Dutta, P., Enhanced uptake and metabolism of riboflavin in erythrocytes infected with Plasmodium falciparum, J. Protozool., 38, 479, 1991.

    123. Mohan, K., Dubey, M. L., Ganguly, N. K., and Mahajan, R. C., Plasmodium falci- parum-induced perturbations of the erythro- cyte anti-oxidant system, Clin. Chim. Acta, 209, 19, 1992.

    124. Mohan, K., Dubey, M. L., Ganguly, N. K., Nain, C. K., and Mahajan, R. C., Increased glutathione cycling and vitamin E of P. falciparum-infected erythrocytes fail to pre- vent spontaneous haemolysis, Ind. J. Biochem. Biophys., 31. 476, 1994.

    125. Sarin, K., Kumar, A., Prakash, A., and Sharma, A., Oxidative stress and anti-oxi- dant defence mechanism in Plasmodium viva malaria before and after chloroquine treat- ment, Ind. J. Malariol., 30, 127, 1993.

    126. Jacobson, F. S.,Morgan, R. W., Christman, M. F., and Ames, B. N., An alkyl hydroper- oxide reductase from Salmonella typhimurium involved in the defense of DNA against oxi- dative damage, J. Biol. Chem., 264, 1488, 1989.

    Inhibitory effects of histidine analogues on growth and protein synthesis by Plasmodium falciparum in vitro, Biochem. Pharmacol., 35, 1589, 1986.

    127. Sample, A. K. and C Z U P ~ ~ ~ ~ , c. J., Elimi- nation of hydrogen peroxide by Haemophilus somnus, a catalase-negative pathogen of cattle, Infect. Immun., 59, 2239, 1991.

    118

    119.

    Panton, L. J., Rossan, R. N., Escajadillo, A., Matasumoto, Y., Lee, A. T., Labroo, V. M., Kirk, K. L., Cohen, L. A., Aikawa, M., and Howard, R. J., In vitro and in vivo studies of the effects of halogenated histi- dine analogs on Plasmodium falciparum, Antimicrob. Agents Chemother., 32, 1655, 1988.

    Chevion, M., A site-specific mechanism for free radical induced biological damage: the essential role of redox-active transition met- als, Free Rad. Biol. Med., 5 , 27, 1988.

    128. Miyake, C., Michihata, F., and Asada, K., Scavenging of hydrogen peroxide in prokary- otic and eukaryotic algae: acquisition of ascor- bate peroxidase during the evolution of cyanobacteria, Plant Cell Physiol., 32, 33, 1991.

    129. Page-Sharp, M., Behm, C. A., and Smith, G. D., Tritrichomonas foetus and Trichomo- nas vaginalis: the pattern of inactivation of hydrogenase activity by oxygen and activities of catalase and ascorbate peroxidase, Micro- biology, 142, 207, 1996.

    31 3

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.

  • 130. Morgan, R. W., Christman, M. F., Jacob- son, F. s., Ston, G., and Ames, B. N., Hy- drogen peroxide-inducible proteins in Salmo- nella typhimurium overlap with heat shock and other stress proteins, Proc. Natl. Acad. Sci. U.S.A., 83, 8059, 1986.

    131. Greenberg, J. T. and Demple, B., A global response induced in Escherichia coli by re- dox-cycling agents overlaps with that induced by peroxide stress, J. Bacteriol., 171, 3933, 1989.

    132. Demple, B. and Amabile-Cuevas, C. F., Redox redux: the control of oxidative stress responses, Cell, 67, 837, 1991.

    133. Albertini, R., Rindi, S., Passi, A., Bardoni, A., Salvini, R., Pallavicini, G., and De Luca, G., The effect of cornea proteoglycans on liposome peroxidase, Arch. Biochem. Bio- phys., 327, 209, 1996.

    134. Bracha, R. and Mirelman, D., Virulence of Entamoeba histolytica trophozoites: effects of bacteria, microaerobic conditions and met- ronidazole, J. Exp. Med., 160, 353, 1984.

    135. Mehlotra, R. K., Entamoeba histolytica: from intestine to liver, Parasitol. Today, 4, 235, 1988.

    136. Kumar, S., Tripathi, L. M., and Sagar, P., Oxido-reductive functions of Entamoeba histolytica in relation to virulence, Ann. Trop. Med. Parasitol., 86, 239, 1992.

    137. McConville, M. J. and Ferguson, M. A. J., The structure, biosynthesis and function of glycosylated phosphatidylinositols in the para- sitic protozoa and higher eukaryotes, Biochem. J., 294, 305, 1993.

    138. Ferguson, M. A. J., Brimacornbe, J. S., Cottaz, S., Field, R. A., Guther, L.S., Homans, S. W., McConville, M. J., Mehlert, A., Milne, K. G., Ralton, J. E.,

    Roy, Y. A., Schneider, P., and Zitzmann, N., Glycosyl-phosphatidylinositol molecules of the parasite and the host, Parasitology, 108, S45, 1994.

    139. Ortner, S., Plaimauer, B., Binder, M., Wiedermann, G., Scheiner, O., and Duchene, M., Humoral immune response against a 70-kilodalton heat shock protein of Entamoeba histolytica in a group of patients with invasive amoebiasis, Mol. Biochem. Parasitol., 54, 175, 1992.

    140. Char, S., Cevallos, A. M., and Farthing, M. J., An immunodominant antigen of Giar- dia lamblia is a heat shock protein, Biotechnol. Ther., 3, 151, 1992.

    141. Requena, J. M., Jimenez-Ruiz, A., Soto, M., Assiego, R., Santaren, J. F., Lopez, M. C., Patarroyo, M. E., and Alonso, C., Regulation of hsp70 expression in Trypano- soma cruzi by temperature and growth phase, Mol. Biochem. Parasitol., 53, 201, 1992.

    142. Kumar, N , Nagasawa, H., Sacci, J. B., Jr., Sina, B. J., Aikawa, M., Atkinson, C., Uparanukraw, P., Kubiak, L.B., Azad, A. F., and Hollingdale, M. R., Expression of members of the heat shock protein 70 family in the exoerythrocytic stages of Plasmodium berghei and Plasmodium falciparum, Paras- itol. Res., 79, 109, 1993.

    143. Biswas, S. and Sharma, Y. D., Enhanced expression of Plasmodium falciparum heat shock protein PFWSP70-I at higher tempera- tures and parasite survival, FEMS Microbiol. Lett., 124, 425, 1994.

    144. Bonnefoy, S., Attal, G., Langsley, G., Tekaia, F., and Mercereau-Puijalon, O., Molecular characterization of the heat shock protein 90 gene of the human malaria parasite Plasmodium falciparum, Mol. Biochem. Parasitol., 67, 157, 1994.

    314

    Criti

    cal R

    evie

    ws i

    n M

    icro

    biol

    ogy

    Dow

    nloa

    ded

    from

    info

    rmah

    ealth

    care

    .com

    by

    Uni

    vers

    ity o

    f Brit

    ish C

    olum

    bia

    on 0

    4/29

    /13

    For p

    erso

    nal u

    se o

    nly.