Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Antimicrob Agents Chemother ; 51(5): 1753-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17307975

ABSTRACT

The ever evolving resistance of the most virulent malaria parasite, Plasmodium falciparum, to antimalarials necessitates the continuous development of new drugs. Our previous analysis of the antimalarial activities of the hemolytic antimicrobial peptides dermaseptins and their acylated derivatives implicated the importance of hydrophobicity and charge for drug action. Following these findings, an oligoacyllysine (OAK) tetramer designed to mimic the characteristics of dermaseptin was synthesized and assessed for its antimalarial activity in cultures of P. falciparum. The tetramer inhibited the growth of different plasmodial strains at low micromolar concentrations (mean 50% inhibitory concentration [IC(50)], 1.8 microM). A structure-activity relationship study involving eight derivatives unraveled smaller, more potent OAK analogs (IC(50)s, 0.08 to 0.14 microM). The most potent analogs were the most selective, with selectivity ratios of 3 orders of magnitude. Selectivity was strongly influenced by the self-assembly properties resulting from interactions between hydrophobic OAKs, as has been observed with conventional antimicrobial peptides. Further investigations performed with a representative OAK revealed that the ring and trophozoite stages of the parasite developmental cycle were equally sensitive to the compound. A shortcoming of the tested compound was the need for long incubation times in order for it to exert its full effect. Nevertheless, the encouraging results obtained in this study regarding the efficiency and selectivity of some compounds establish them as leads for further development.


Subject(s)
Antimalarials/pharmacology , Lysine/analogs & derivatives , Peptides/pharmacology , Plasmodium falciparum/drug effects , Amino Acid Sequence , Animals , Cell Line , Dogs , Hemolysis/drug effects , Molecular Sequence Data , Peptides/chemistry , Peptides/toxicity , Structure-Activity Relationship
2.
Mol Biochem Parasitol ; 117(2): 161-8, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11606226

ABSTRACT

Transglutaminase was identified in malaria parasites by immunofluorescence microscopy using alpha-transglutaminase antiserum. Functional enzyme was demonstrated in vivo and in vitro using labeled polyamines that become incorporated into protein substrates through TGase activity. In Plasmodium falciparum intraerythrocytic parasites, transglutaminase activity was stage-dependent: it was weak in ring-forms but much stronger in trophozoites and schizonts. High levels of activity were detected in P. gallinaceum zygotes and ookinetes and in capsules of oocysts developing on mosquito midguts. Unlike most known transglutaminases, the enzymatic activity in Plasmodium was Ca(2+)-independent. Furthermore, levels of activity were similar at 37 and 26 degrees C. Parasite transglutaminase may be responsible for the modification of erythrocytic cytoskeleton in infected cells and it may facilitate the construction of oocyst capsules by cross-linking mosquito-derived basement membrane components with Plasmodium-derived proteins.


Subject(s)
Chickens/parasitology , Plasmodium falciparum/enzymology , Plasmodium falciparum/growth & development , Plasmodium gallinaceum/enzymology , Plasmodium gallinaceum/growth & development , Transglutaminases/metabolism , Aedes/parasitology , Animals , Calcium/metabolism , Erythrocytes/parasitology , Humans , Malaria, Avian/parasitology , Malaria, Falciparum/parasitology , Transglutaminases/antagonists & inhibitors
3.
Antimicrob Agents Chemother ; 44(9): 2442-51, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10952593

ABSTRACT

The hemolytic antimicrobial peptide dermaseptin S4 was recently shown to exert antimalarial activity. In this study, we attempted to understand the underlying mechanism(s) and identify derivatives with improved antimalarial activity. A number of dermaseptin S4 derivatives inhibited parasite growth with a 50% inhibitory concentration (IC(50)) in the micromolar range. Among these, the substituted S4 analog K(4)K(20)-S4 was the most potent (IC(50) = 0.2 microM), while its shorter version, K(4)-S4(1-13)a, retained a considerable potency (IC(50) = 6 microM). Both K(4)K(20)-S4 and K(4)-S4(1-13)a inhibited growth of the parasites more at the trophozoite stage than at the ring stage. Significant growth inhibition was observed after as little as 1 min of exposure to peptides and proceeded with nearly linear kinetics. The peptides selectively lysed infected red blood cells (RBC) while having a weaker effect on noninfected RBC. Thus, K(4)K(20)-S4 lysed trophozoites at concentrations similar to those that inhibited their proliferation, but trophozoites were >30-fold more susceptible than normal RBC to the lytic effect of K(4)K(20)-S4, the most hemolytic dermaseptin. The same trend was observed with K(4)-S4(1-13)a. The D isomers of K(4)K(20)-S4 or K(4)-S4(1-13)a were as active as the L counterparts, indicating that antimalarial activity of these peptides, like their membrane-lytic activity, is not mediated by specific interactions with a chiral center. Moreover, dissipation of transmembrane potential experiments with infected cells indicated that the peptides induce damage in the parasite's plasma membrane. Fluorescence confocal microscopy analysis of treated infected cells also indicated that the peptide is able to find its way through the complex series of membranes and interact directly with the intracellular parasite. Overall, the data showed that dermaseptins exert antimalarial activity by lysis of infected cells. Dermaseptin derivatives are also able to disrupt the parasite plasma membrane without harming that of the host RBC.


Subject(s)
Amphibian Proteins , Antimalarials/pharmacology , Antimicrobial Cationic Peptides , Peptides/pharmacology , Plasmodium falciparum/drug effects , Animals , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Antimalarials/chemistry , Cell Membrane/drug effects , Cell Membrane/physiology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Kinetics , Membrane Potentials/drug effects , Microscopy, Confocal , Peptides/chemistry , Plasmodium falciparum/physiology , Stereoisomerism
4.
Proc Natl Acad Sci U S A ; 97(1): 331-6, 2000 Jan 04.
Article in English | MEDLINE | ID: mdl-10618418

ABSTRACT

The increasing resistance of the malaria parasite Plasmodium falciparum to currently available drugs demands a continuous effort to develop new antimalarial agents. In this quest, the identification of antimalarial effects of drugs already in use for other therapies represents an attractive approach with potentially rapid clinical application. We have found that the extensively used antimycotic drug clotrimazole (CLT) effectively and rapidly inhibited parasite growth in five different strains of P. falciparum, in vitro, irrespective of their chloroquine sensitivity. The concentrations for 50% inhibition (IC(50)), assessed by parasite incorporation of [(3)H]hypoxanthine, were between 0.2 and 1.1 microM. CLT concentrations of 2 microM and above caused a sharp decline in parasitemia, complete inhibition of parasite replication, and destruction of parasites and host cells within a single intraerythrocytic asexual cycle (approximately 48 hr). These concentrations are within the plasma levels known to be attained in humans after oral administration of the drug. The effects were associated with distinct morphological changes. Transient exposure of ring-stage parasites to 2.5 microM CLT for a period of 12 hr caused a delay in development in a fraction of parasites that reverted to normal after drug removal; 24-hr exposure to the same concentration caused total destruction of parasites and parasitized cells. Chloroquine antagonized the effects of CLT whereas mefloquine was synergistic. The present study suggests that CLT holds much promise as an antimalarial agent and that it is suitable for a clinical study in P. falciparum malaria.


Subject(s)
Antimalarials/pharmacology , Clotrimazole/pharmacology , Plasmodium falciparum/drug effects , Animals , Antifungal Agents/pharmacology , Cells, Cultured , Chloroquine/pharmacology , Growth Inhibitors/pharmacology , Histocytochemistry , Hypoxanthine/metabolism , Mefloquine/pharmacology , Plasmodium falciparum/growth & development , Plasmodium falciparum/parasitology
5.
Biochem Pharmacol ; 58(1): 59-68, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10403519

ABSTRACT

We have shown previously that chloroquine and amodiaquine inhibit the glutathione-dependent degradation of ferriprotoporphyrin IX (FP). We have also demonstrated that treatment of human erythrocytes infected with Plasmodium falciparum with chloroquine or amodiaquine results in a dose- and time-dependent accumulation of FP in the membrane fraction of these cells in correlation with parasite killing. High levels of membrane FP are known to perturb the barrier properties of cellular membranes, and could thereby irreversibly disturb the ion homeostasis of the parasite and cause parasite death. We here report on the effect of various 4-aminoquinolines, as well as pyronaridine, halofantrine and some bis-quinolines, on glutathione-mediated destruction of FP in aqueous solution, when FP was bound non-specifically to a protein, and when it was dissolved in human erythrocyte ghost membranes. We showed that all drugs were capable of inhibiting FP degradation in solution. The inhibitory efficacy of some drugs declined when FP was bound non-specifically to protein. Quinine and mefloquine were unable to inhibit the degradation of membrane-associated FP, in line with their inability to increase membrane-associated FP levels in malaria-infected cells following drug treatment. The discrepancy between chloroquine and amodiaquine on the one hand, and quinine and mefloquine on the other, is discussed in terms of the particular location of drugs and FP in the phospholipid membrane, and may suggest differences in the mechanistic details of the antimalarial action of these drugs.


Subject(s)
Amodiaquine/pharmacology , Antimalarials/pharmacology , Chloroquine/pharmacology , Erythrocytes/drug effects , Glutathione/metabolism , Hemin/metabolism , Animals , Dose-Response Relationship, Drug , Erythrocyte Membrane/drug effects , Erythrocyte Membrane/metabolism , Erythrocytes/metabolism , Humans , In Vitro Techniques , Kinetics , Plasmodium falciparum/drug effects , Plasmodium falciparum/metabolism
6.
Mol Biochem Parasitol ; 99(1): 129-41, 1999 Mar 15.
Article in English | MEDLINE | ID: mdl-10215030

ABSTRACT

The intraerythrocytic malaria parasite digests considerable amounts of its host cell cytosol, which consists mostly of hemoglobin. In order to avert the toxicity of ferriprotorphyrin IX (FP) thus produced, it is generally accepted that FP is polymerized to the non-toxic hemozoin. Investigating the fate of FP in cultured Plasmodium falciparum -infected human red blood cells, revealed a straight correlation between amounts of digested hemoglobin and hemozoin, but the latter contained less FP than produced. The efficacy of FP polymerization is stage-dependent, increasing with parasite maturation. Different strains display dissimilar efficacy in hemozoin production. Unpolymerized FP possibly exits the food vacuole and is degraded by glutathione, thus accounting for the low levels of free FP found in infected cells. 4-aminoquinoline antimalarials demonstrably form complexes with FP and inhibit hemozoin production in vitro. Chloroquine, amodiaquine, quinine and mefloquine were found to inhibit hemozoin production in intact infected cells, but only the first two drugs caused a dose-dependent accumulation of FP in the membrane fraction of infected cells that correlated well with parasite killing, due to the permeabilization of membranes to ions. This differential effect is explained by the ability of chloroquine and amodiaquine to inhibit the degradation of membrane-associated FP by glutathione and the incapacity of quinine and mefloquine to do so. This discrepancy implies that the antimalarial mode of action of chloroquine and amodiaquine is different in its mechanistic details from that of quinine and mefloquine and is compatible with the diametric sensitivity of most strains to chloroquine and mefloquine and the disparate interaction of these drugs with enhancers of their antimalarial action.


Subject(s)
Antimalarials/pharmacology , Erythrocytes/parasitology , Hemin/metabolism , Plasmodium falciparum/drug effects , Plasmodium falciparum/metabolism , Animals , Cells, Cultured , Hemeproteins/metabolism , Hemoglobins/metabolism , Humans , Plasmodium falciparum/growth & development
7.
Biochem Pharmacol ; 56(10): 1305-13, 1998 Nov 15.
Article in English | MEDLINE | ID: mdl-9825729

ABSTRACT

We propose here a new and detailed model for the antimalarial action of chloroquine (CQ), based on the its ability to inhibit degradation of heme by glutathione. Heme, which is toxic to the malaria parasite, is formed when the intraerythrocytic malaria parasite ingests and digests inside its food vacuole its host cell cytosol, which consists mainly of hemoglobin. The parasite protects itself against the toxicity of heme by polymerizing some of it to insoluble hemozoin (HZ). We show here that in Plasmodium falciparum at the trophozoite stage only ca. 30% of the heme is converted into hemozoin. We suggest that nonpolymerized heme exits the food vacuole and is subsequently degraded by glutathione, as has been shown before for uninfected erythrocytes. Marginal amounts of free heme could be detected in the membrane fraction of infected cells but nowhere else. It is well established that CQ and amodiaquine (AQ) accumulate in the parasite's food vacuole and inhibit heme polymerization, thereby increasing its efflux out of the food vacuole. We found that these drugs competitively inhibit the degradation of heme by glutathione, thus allowing heme to accumulate in membranes. Incubation of intact infected cells with CQ and AQ results in a marked increase in membrane-associated heme in a dose- and time-dependent manner, and a relationship exists between membrane heme levels and the extent of parasite killing. Heme has been shown to disrupt the barrier properties of membranes and to upset ion homeostasis in CQ-treated malaria-infected cells. In agreement with the predictions of our model, increasing the cellular levels of glutathione leads to increased resistance to CQ, whereas decreasing them results in enhanced sensitivity to the drug. These results insinuate a novel mechanism of drug resistance.


Subject(s)
Amodiaquine/pharmacology , Antimalarials/pharmacology , Chloroquine/pharmacology , Glutathione/antagonists & inhibitors , Heme/metabolism , Animals , Glutathione/metabolism , Plasmodium falciparum/drug effects
8.
Trans R Soc Trop Med Hyg ; 90(5): 558-62, 1996.
Article in English | MEDLINE | ID: mdl-8944274

ABSTRACT

Phagocytic cells constitute the first line of defence against malarial parasites. They perform their role by delivering oxidative radicals and by phagocytosing infected red blood cells (IRBC). Phagocytosis is mediated by antibody binding to clustered band 3 antigen in the IRBC membrane and activation of the alternative complement pathway. In this study we showed that treatment of IRBC containing Plasmodium falciparum with therapeutically-relevant concentrations of antimalarial drugs considerably reduced the binding of immunoglobulin G (IgG) to, and the phagocytosis of, IRBC. Opsonization of IRBC by fresh serum before drug treatment prevented this inhibitory action of drugs. Removal of the drug restored IgG binding and the phagocytic susceptibility of IRBC in a time-dependent fashion. Direct measurement of the effect of chloroquine on the clustering of band 3 in IRBC, however, failed to reveal any disruption of the aggregation. We conclude that antimalarial drugs are able to alter, by an as yet unresolved mechanism, the affinity of IgG to clustered band 3. This affinity of IRBC seems to be determined by a dynamic process that depends on the metabolic activity of the parasite.


Subject(s)
Antimalarials/pharmacology , Malaria, Falciparum/drug therapy , Antibody Affinity/drug effects , Autoantibodies/immunology , Erythrocytes/immunology , Erythrocytes/parasitology , Humans , Immunoglobulin G , In Vitro Techniques , Monocytes/drug effects , Opsonin Proteins , Phagocytosis/drug effects , Time Factors
9.
Parasitology ; 113 ( Pt 1): 7-18, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8710417

ABSTRACT

The balanced polymorphism of glucose-6-phosphate dehydrogenase deficiency (G6PD-) is believed to have evolved through the selective pressure of malarial combined with consumption of fava beans. The implicated fava bean constituents are the hydroxypyrimidine glucosides vicine and convicine, which upon hydrolysis of their beta-O-glucosidic bond, became protein pro-oxidants. In this work we show that the glucosides inhibit the growth of Plasmodium falciparum, increase the hexose-monophosphate shunt activity and the phagocytosis of malaria-infected erythrocytes. These activities are exacerbated in the presence of beta-glucosidase, implicating their pro-oxidant aglycones in the toxic effect, and are more pronounced in infected G6PD- erythrocytes. These results suggest that G6PD- infected erythrocytes are more susceptible to phagocytic cells, and that fava bean pro-oxidants are more efficiently suppressing parasite propagation in G6PD- erythrocytes, either by directly affecting parasite growth, or by means of enhanced phagocytic elimination of infected cells. The present findings could account for the relative resistance of G6PD- bearers to falciparum malaria, and establish a link between dietary habits and malaria in the selection of the G6PD- genotype.


Subject(s)
Glucosephosphate Dehydrogenase Deficiency/complications , Glucosides/pharmacology , Malaria, Falciparum/complications , Phagocytosis/drug effects , Plasmodium falciparum/drug effects , Pyrimidinones/pharmacology , Uridine/analogs & derivatives , Animals , Erythrocytes/drug effects , Erythrocytes/enzymology , Erythrocytes/parasitology , Fabaceae , Female , Glucosephosphate Dehydrogenase/blood , Glucosephosphate Dehydrogenase Deficiency/genetics , Glucosides/metabolism , Humans , Hydrogen-Ion Concentration , Hydrolysis , Male , Pentose Phosphate Pathway/drug effects , Plants, Medicinal , Plasmodium falciparum/growth & development , Plasmodium falciparum/immunology , Pyrimidinones/metabolism , Uridine/metabolism , Uridine/pharmacology
10.
Biochem Pharmacol ; 51(5): 693-700, 1996 Mar 08.
Article in English | MEDLINE | ID: mdl-8615907

ABSTRACT

The antimalarial action of methylene blue (MB) was first noted by Paul Ehrlich in the late 19th century. Although it has only sporadically been adopted as a serviceable drug, the resolution of its antimalarial action seems warranted, as it is currently used for the treatment of various methemoglobinemias. In this work we have used MB, and its analogues Azures A (AZA), B (AZB), C (AZC), and thionin (TH), as well as the oxazine Celestine blue (CB) and azine Phenosaphranin (PS). All MB analogues inhibit the growth of various strains of Plasmodium falciparum in culture with IC50s in the 2 x 10(-9)-1 x 10(-7) M range, with the rank order MB approximately AZA > AZB > AZC > TH > PS > CB. The IC50s for a mammalian cell line were in the 3 x 10(-6)-4 x 10(-5) M range, and the rank order was TH approximately AZB > AZA approximately PS > AZC approximately CB > MB. As MB could affect cell growth through the oxidation of NADPH, we tested the action of the various compounds on the hexose-monophosphate shunt activity. Appreciable activation of the shunt was observed at 1 x 10(-5) M in both cell types, thus accounting for inhibition of growth of mammalian cells but not of parasites. All compounds were found to complex with heme in a rank order similar to their antimalarial effect. It is therefore suggested that MB and its congeners act by preventing the polymerization of heme, which is produced during the digestion of host cell cytosol in the parasite food vacuole, into hemozoin. In this respect, these compounds seem to act similarly to the 4-aminoquinoline antimalarials. All compounds effectively suppressed the growth of P. vinckei petteri in vivo with IC50 in the 1.2-5.2 mg/kg range, and MB and AZB suppressed P. yoelii nigeriensis in the 9-11 mg/kg range (i.e. at doses similar to those of chloroquine). The potential toxicity of these compounds may restrict their clinical use, but their impressive antimalarial activities suggest that the phenothiazine structure could serve as a lead compound for further drug development.


Subject(s)
Antimalarials/pharmacology , Malaria/drug therapy , Methylene Blue/pharmacology , Plasmodium falciparum/drug effects , Plasmodium yoelii/drug effects , Animals , Heme/metabolism , Humans , Male , Methylene Blue/metabolism , Mice , Oxidation-Reduction , Pentose Phosphate Pathway/drug effects , Tumor Cells, Cultured
11.
Int J Parasitol ; 25(12): 1457-62, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8719958

ABSTRACT

Intralipid and Ivelip are commercial preparations of soy-bean lipid extracts used for intravenous supplementation of lipids in various clinical conditions. They were found to inhibit the growth of Plasmodium falciparum in culture with an IC50 of 8.07 +/- 2.13 and 13.32 +/- 2.05 mg.ml-1, respectively. Intralipid rapidly and efficiently inhibited nucleic acid synthesis in cultured P. falciparum, exhibiting full inhibitory activity in less than 2 h. Ivelip injected intraperitoneally, was found by the 4-day suppressive test to be active in vivo against P. vinckei petteri within the normal recommended regimen for dietary lipid supply (0.5-4 g.kg-1), but it was impossible to obtain a radical cure even with very high doses (6.4 g.kg-1). Ivelip was less effective against P. berghei and P. yoelii nigeriensis. As Ivelip showed no interference with the antimalarial activity of chloroquine, it could be considered for use in the treatment of severe human malaria in association with 4-aminoquinolines to expedite the clearance of parasites.


Subject(s)
Antimalarials/pharmacology , Fat Emulsions, Intravenous/pharmacology , Plasmodium/drug effects , Animals , Antimalarials/administration & dosage , Chloroquine/administration & dosage , Fat Emulsions, Intravenous/administration & dosage , Female , Humans , Malaria/drug therapy , Malaria/parasitology , Mice , Nucleic Acids/biosynthesis , Oxidative Stress , Plasmodium/growth & development , Plasmodium/metabolism , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects , Plasmodium yoelii/drug effects , Glycine max
12.
Exp Parasitol ; 81(1): 97-105, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7628573

ABSTRACT

Following the demonstration of the antimalarial effect of the long chain saturated alcohol n-hentriacontanol ((CH2)29CH2OH), isolated from the Bolivian endemic solanaceous plant Cuatresia sp., we have tested the effect of the C18 fatty acids oleic, elaidic, linoleic, and linoleic on malaria parasites. These fatty acids inhibited the parasitemic development in mice infected with Plasmodium vinckei petteri or with Plasmodium yoelii nigeriensis in a 4-day suppressive test. To gain a deeper discernment of the antimalarial mode of action, the effects of these compounds were evaluated on Plasmodium falciparum growth in culture. Whereas n-hentriacontanol did not show any inhibition of this parasite, on the contrary, the C18 acids displayed a considerably inhibitory activity at < or = 200 micrograms/ml both in intact infected cells and in free parasites. In order to understand the mechanism of their antimalarial action, several tests were performed. No hemolysis of infected cells could be observed up to 500 microgram/ml. No effect on the lipid peroxidation, ATP levels, transport through the parasite-induced permeability pathways, or on the phagocytosis of the infected cells could be observed. The cytotoxic effect of the fatty acids was very rapid: full inhibition of nucleic acids and protein syntheses was observed in less than 30 min. This inhibition was not relieved by the addition of deferrioxamine or FeCl3, indicating that fatty acids (FA) do not act by facilitating the transport of iron. Inhibition was relieved in neither the presence of orotic acid or its methyl ester, indicating that FA do not act at the mitochondrial level of pyrimidine synthesis.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antimalarials/pharmacology , Antimalarials/therapeutic use , Fatty Acids, Nonesterified/pharmacology , Fatty Acids, Nonesterified/therapeutic use , Malaria/drug therapy , Plasmodium falciparum/drug effects , Plasmodium yoelii/drug effects , Plasmodium/drug effects , Animals , Linoleic Acid , Linoleic Acids/pharmacology , Linoleic Acids/therapeutic use , Male , Mice , Oleic Acid , Oleic Acids/pharmacology , Oleic Acids/therapeutic use , Parasitemia/prevention & control , Structure-Activity Relationship , alpha-Linolenic Acid/pharmacology , alpha-Linolenic Acid/therapeutic use
13.
Parasitology ; 108 ( Pt 4): 371-81, 1994 May.
Article in English | MEDLINE | ID: mdl-8008450

ABSTRACT

Intraerythrocytic malaria parasites ingest the cytosol of their host cell and digest it inside their acid food vacuoles. Acidified (pH 4-5.5, 37 degrees C) human red blood cell lysates were used to simulate this process, measuring the denaturation of haemoglobin (Hb) and the release of iron, in the absence or presence of exogenous protease. Spontaneous Hb denaturation and appearance of non-heme iron were observed upon lysate acidification, their rates decreasing with increasing pH, and increasing in the presence of protease. Both processes were inhibited by the quinoline-containing anti-malarial drugs (QCDs) chloroquine, quinine, mefloquine and amodiaquine at concentrations well below those expected in the acidic food vacuole of the parasite. Spectrophotometric analysis indicated that chloroquine complexes with heme in acid-denatured haemoglobin. Other weak bases as well as verapamil and diltiazem, known to reverse the resistance of malarial parasites to chloroquine, were without effect indicating that the action of QCDs is specific. Based on our previous results and the present report, we suggest that iron release in acidified lysates is mediated through the formation of ferryl (Fe(IV)) radicals. QCDs possibly complex with this radical, as they do with heme, and prevent its contact with an adjacent heme molecule which is required for ring opening and iron release. These results may suggest that one of the anti-malarial effects of QCDs is to deprive the parasite of an adequate iron supply. Addition of iron to cultures of Plasmodium falciparum was expected to circumvent the deprivation of iron and reduce the anti-malarial effect of QCDs. However, adding iron as penetrating fructose or nitrilotriacetate complexes did not alter the parasite's susceptibility to chloroquine. Ascorbate markedly increased the release of iron in acidified lysates, and this effect was not reduced by chloroquine. Ascorbate was found to decrease parasite susceptibility to chloroquine, suggesting that iron deprivation may be an important factor in the anti-malarial action of QCDs.


Subject(s)
Antimalarials/pharmacology , Erythrocytes/drug effects , Hemoglobins/drug effects , Iron/metabolism , Plasmodium falciparum/drug effects , Quinolines/pharmacology , Animals , Ascorbic Acid/pharmacology , Diltiazem/pharmacology , Dose-Response Relationship, Drug , Erythrocytes/metabolism , Heme/metabolism , Hemoglobins/metabolism , Humans , Hydrogen-Ion Concentration , Verapamil/pharmacology
14.
Mol Biochem Parasitol ; 58(1): 7-15, 1993 Mar.
Article in English | MEDLINE | ID: mdl-7681547

ABSTRACT

The DNA of malarial parasites is significantly richer in A and T than that of mammalian cells. Antibiotics which bind to the minor groove of B-DNA with a preference for AT-rich sequences, such as distamycin A, netropsin, 4'-6-diamidino-2-phenylindole (DAPI) and bis-benzimide (Hoechst 33258) were found to inhibit the growth and propagation of Plasmodium falciparum in culture. Distamycin A readily inhibited nucleic acid and protein synthesis and was more toxic to the ring stage than to the trophozoite stage in various parasite strains, irrespective of their susceptibility to chloroquine. Distamycin A, netropsin, DAPI and Hoechst 33258 were considerably more toxic to parasites than to mammalian cells, while chromomycin A3 and mithramycin A, which bind preferentially to GC-rich sequences, were either equally toxic or more harmful to mammalian cells. These results suggest that the mere difference in DNA base composition of parasites and host cells may account for the selective toxicity of minor groove ligands. Distamycin A, DAPI and Hoechst 33258 were also found to be more toxic to Saccharomyces cerevisiae grown on glycerol than to yeast cells grown on glucose, consistent with the preferential binding of these ligands to the relatively AT-rich mitochondrial DNA of yeast cell. These results underscore the generality of selective toxicity of minor groove binders endowed by the DNA base composition.


Subject(s)
DNA, Protozoan/drug effects , Intercalating Agents/pharmacology , Plasmodium falciparum/drug effects , Animals , Base Composition , Bisbenzimidazole/pharmacology , Cell Division/drug effects , Cells, Cultured , Chromomycins/pharmacology , Distamycins/pharmacology , Dose-Response Relationship, Drug , Humans , Indoles/pharmacology , Mice , Netropsin/pharmacology , Nucleic Acid Conformation , Plasmodium falciparum/growth & development , Plicamycin/analogs & derivatives , Plicamycin/pharmacology , Saccharomyces cerevisiae/drug effects
15.
Biochem Pharmacol ; 43(1): 63-70, 1992 Jan 09.
Article in English | MEDLINE | ID: mdl-1734899

ABSTRACT

Malaria constitutes one of the major health threats in the tropical and sub-tropical areas of the world. Yet, few advances were made in recent years in revealing the mode of action of the common and most economically affordable antimalarial drugs, the schizontocidal 4-aminoquinolines. Data presented indubitably repudiate the previous notions that these drugs act by either halting the feeding of the parasite on its host erythrocyte cytosol or repressing nucleic acid synthesis due to intercalation into the parasite's DNA. A novel target for drugs is outlined, i.e. they are shown to inhibit in vitro the release of iron from acidified host cell cytosol, consisting mostly of hemoglobin, a process that could provide this trace element to the parasite. Resistance to quinoline-containing drugs is the principal reason for the present resurgence of malaria. Drug-resistant parasites accumulate less of these weak base-like drugs in the acidic digestive vacuoles. A kinetic model is presented, indicating that diminishing drug accumulation is due to decreased vacuolar proton pump activity and is not a result of a putative multidrug resistance (MDR) efflux pump. Findings to date on the molecular biology of parasite mdr genes are reviewed. These indicate no correlation between gene expression or mutations and phenotypic drug resistance. Reversal of parasite drug resistance by relevant compounds in MDR cancer cells seems to involve mechanism(s) different from the inhibition of the MDR pump in cancer cells.


Subject(s)
Antimalarials/pharmacology , Plasmodium falciparum/drug effects , Quinolines/pharmacology , Animals , Antimalarials/metabolism , Drug Resistance/genetics , Intercalating Agents/pharmacology , Plasmodium falciparum/genetics , Quinolines/metabolism
16.
Life Sci ; 49(17): 1213-9, 1991.
Article in English | MEDLINE | ID: mdl-1943436

ABSTRACT

The quinoline-containing antimalarial drugs chloroquine, quinine and mefloquine exert an irreversible inhibitory effect on erythrocytic stages of Plasmodium falciparum grown in culture. Inhibition is time- and concentration-dependent and the full effect is observed after 2-6 hours of exposure to the drug. Washing of infected cells after drug exposure in the presence of NH4Cl to accelerate drug efflux, intensifies the inhibitory effect of chloroquine, probably due to the pH-dependent release of highly concentrated drug from the acidic food vacuole of the parasite. When both antimalarials and NH4Cl are present in the culture, drug effect is reduced, as expected from the demonstrable alkalinization of the food vacuole and the consequent reduction in drug accumulation. The protease inhibitor leupeptin inhibits digestion of ingested host cell cytosol, and thus inhibits parasite growth, though reversibly so (Rosenthal et al, J. Clin. Invest. 82 1560-1566 (1988)). Thus, although the antimalarials also inhibit the feeding process, this is not the cause of their irreversible action. Leupeptin is found to be antagonistic to antimalarials' action, suggesting that the drugs form complexes with products of host cell digestion that are responsible for irreversible inhibition of parasite growth.


Subject(s)
Ammonium Chloride/pharmacology , Antimalarials/pharmacology , Leupeptins/pharmacology , Plasmodium falciparum/drug effects , Quinolines/pharmacology , Animals , Antimalarials/antagonists & inhibitors , Drug Combinations , Drug Interactions , Hydrogen-Ion Concentration , Quinolines/antagonists & inhibitors , Time Factors
17.
Biochem Pharmacol ; 38(16): 2645-54, 1989 Aug 15.
Article in English | MEDLINE | ID: mdl-2669763

ABSTRACT

Quinoline-containing antimalarial drugs accumulate inside the acid food vacuole of the parasite where they inhibit the digestion of ingested host cell cytosol, and consequently, parasite growth. In order to verify whether this inhibition is caused by drug-induced alkalinization of the food vacuole, we investigated the accumulation of acridine orange (AO) as a vacuolar pH probe in intact Plasmodium falciparum-infected human erythrocytes as affected by the drugs chloroquine (CQ), 7H-quinoleine (7HQ), quinine (Q) and mefloquine (MQ). It was established by various criteria that AO accumulates primarily in the acid compartment(s) of the parasite as a function of the pH difference between it and the extracellular medium. This pH gradient was dissipated by the drugs in the rank order MQ greater than CQ greater than Q greater than 7HQ. The kinetics of vacuolar alkalinization and the concentration ranges at which it was observed imply that the monoprotic drugs MQ and Q exerted their effect mostly by translocating protons across the vacuolar membrane, i.e. they could cross the membrane as a protonated species, while the diprotic drugs CQ and 7HQ raised the vacuolar pH mostly by proton trapping. Similarly, hydrophobic alkylamines raised the vacuolar pH by proton translocation, while their relatively more polar congeners and ammonia did so by proton titration. However, the alkalinizing effect of each drug was observed at a concentration which was 1-2 orders of magnitude larger than the IC50 of its antimalarial effect. These results mean that vacuolar alkalinization is not the primary effect of antiparasitic action of quinoline antimalarials.


Subject(s)
Amines/pharmacology , Antimalarials/pharmacology , Erythrocytes/drug effects , Plasmodium falciparum/drug effects , Quinolines/pharmacology , Acridine Orange/pharmacology , Animals , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Erythrocytes/parasitology , Ethylmaleimide/pharmacology , Humans , Hydrogen-Ion Concentration , In Vitro Techniques , Vacuoles/drug effects
18.
Biochem Pharmacol ; 38(8): 1271-7, 1989 Apr 15.
Article in English | MEDLINE | ID: mdl-2650686

ABSTRACT

Various lysosomotropic detergents were tested in this work on in vitro cultures of Plasmodium falciparum and are shown to be potent antimalarial agents. The order of antimalarial potency was similar to that of cell toxicity on mammalian cells in culture (Miller DK et al., J Cell Biol 97, 1841-51 (1983]. The most efficient agents, N-dodecyl-imidazole (NDI) and N-dodecyl morpholine (NDM) displayed IC50 values of 6.7 +/- 0.7 microM and 23 +/- 5 microM. The mechanism of action of NDI measured at IC50 concentrations displayed the following features: irreversible antimalarial effect after 15 min exposure of cells to drug; alkalinization of the parasite food vacuole; inhibition of protein synthesis; inhibition of host cell protein digestion by the parasite; lack of vacuolar membrane disruption; lack of effect on the rate of constitutive autoproteolysis. No biochemical or ultrastructural indications were found to support a detergent-like action of NDI and its structural congeners on the major acidic compartment of the parasite, the food vacuole. Rather, alkalinization of that compartment by weak-base accumulation properties of the amphiphilic drugs and ensuing protonophoric effect are likely to play a major role in the various parasite-associated properties affected by these drugs.


Subject(s)
Antimalarials , Detergents/pharmacology , Plasmodium falciparum/drug effects , Surface-Active Agents/pharmacology , Ammonium Chloride/pharmacology , Animals , Antigens, Protozoan/biosynthesis , Cytosol/metabolism , Cytosol/parasitology , Erythrocytes/parasitology , Humans , Hydrogen-Ion Concentration , Imidazoles/pharmacology , In Vitro Techniques , Lysosomes/drug effects , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Vacuoles/drug effects
19.
Biochem Pharmacol ; 37(10): 2013-8, 1988 May 15.
Article in English | MEDLINE | ID: mdl-3288209

ABSTRACT

Quinoline-containing antimalarials are cationic amphiphiles which accumulate to high levels in lysosomes and are known to interact with membrane phospholipids. It was therefore hypothesized that they could exert their antimalarial effect by compromising the integrity of the parasite's acidic organelles. To test this hypothesis, the effects of chloroquine (CQ), quinine (Q) and mefloquine (MQ) on the osmotic stability of human red blood cells exposed to hypotonic solutions have been investigated. With CQ and Q stabilization was observed at pH 7.8 and destabilization at pH 5, indicating that destabilization is caused by the protonated forms of the drugs. With MQ the pH dependence was reversed, i.e. it destabilized at pH 7.8 and stabilized at pH 5, suggesting that destabilization is caused by the unprotonated drug. MQ caused cell lysis at the tenth millimolar range by a detergent effect. The possible destabilizing effect of drugs on the membranes of Plasmodium falciparum acidic organelles was investigated in metabolically-labelled parasites. We expected an increase in degradation of parasite proteins if drugs did indeed cause the release of acid hydrolases from destabilized organelles to the cytoplasm. No effect of drugs on parasite protein degradation could be observed, but protein synthesis was inhibited at therapeutic drug concentrations. These results imply that quinoline-containing antimalarials do not compromise the integrity of parasite acidic organelles, and that inhibition of protein synthesis results from a limited supply of essential amino acid(s) due to the demonstrable drug-mediated suppression of parasite digestion of host cell cytosol.


Subject(s)
Antimalarials/pharmacology , Erythrocyte Membrane/drug effects , Plasmodium falciparum/drug effects , Animals , Chloroquine/pharmacology , Hemolysis/drug effects , Humans , Hydrogen-Ion Concentration , Mefloquine , Phospholipids/metabolism , Plasmodium falciparum/metabolism , Potassium/metabolism , Proteins/metabolism , Quinine/pharmacology , Quinolines/pharmacology
20.
Life Sci ; 40(13): 1253-7, 1987 Mar 30.
Article in English | MEDLINE | ID: mdl-3550344

ABSTRACT

Human erythrocytes were loaded with either gentamicin or amikacin and subsequently infected with the human malarial parasite Plasmodium falciparum and grown in culture. Parasite invasion of erythrocytes was unaffected by the drugs, but subsequent development was retarded. The digestion of host cell cytosol in ring-stage parasites was inhibited by the drugs. A substantial acid, Ca2+-independent phospholipase activity could be monitored in parasite cytosol and was found to be inhibited by the drugs. These results imply that phospholipases are involved in the feeding mechanism of the parasite and that gentamicin and amikacin exert their inhibitory activity by affecting these enzymes.


Subject(s)
Amikacin/pharmacology , Antimalarials/pharmacology , Gentamicins/pharmacology , Phospholipases A/antagonists & inhibitors , Phospholipases/antagonists & inhibitors , Plasmodium falciparum/drug effects , Erythrocytes/parasitology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...