You are on page 1of 17

Combinatorial Chemistry & High Throughput Screening, 2005, 8, 63-79 63

Targeting the Hemozoin Synthesis Pathway for New Antimalarial Drug


Discovery: Technologies for In Vitro β-Hematin Formation Assay
Babu L. Tekwani* and Larry A. Walker

National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of
Pharmacy, University of Mississippi, University, MS 38677, USA
Abstract: Clinical manifestations of malaria primarily result from proliferation of the parasite within the
hosts’ erythrocytes. During this process, hemoglobin is utilized as the predominant source of nutrition. The
malaria parasite digests hemoglobin within the digestive vacuole through a sequential metabolic process
involving multiple proteases. Massive degradation of hemoglobin generates large amount of toxic heme.
Malaria parasite, however, has evolved a distinct mechanism for detoxification of heme through its conversion
into an insoluble crystalline pigment, known as hemozoin. Hemozoin is identical to β-hematin, which is
constituted of cyclic heme dimers arranged in an ordered crystalline structure through intermolecular
hydrogen bonding. The exact mechanism of biogenesis of hemozoin in malaria is still obscure and is the
subject of intense debate. Hemozoin synthesis is an indispensable process for the parasite and is the target for
action of several known antimalarials. The pathway has therefore attracted significant interest for new
antimalarial drug discovery research. Formation of β-hematin may be achieved in vitro under specific chemical
and physiochemical conditions through a biocrystallization process. Based on these methods several
experimental approaches have been described for the assay of formation of β-hematin in vitro and screening of
compounds as inhibitors of hemozoin synthesis. These assays are primarily based on differential solubility
and spectral characteristics of monomeric heme and β-hematin. Different factors viz., the malaria parasite lysate,
lipids extracts, preformed β-hematin, malarial histidine rich protein II and some unsaturated lipids have been
employed for promoting β-hematin formation in these assays. The assays based on spectrophotometric
quantification of β-hematin or incorporation of 14 C-heme yield reproducible results and have been applied to
high throughput screening. Several novel antimalarial pharmacophores have been discovered through these
assays.
Keywords: Malaria, Plasmodium falciparum, hemozoin, beta-hematin, heme, antimalarials, quinolines.

1. INTRODUCTION parasite afford several distinct metabolic characteristics and


unique metabolic pathways, which are different from the
For thousand of years malaria has been a major cause of
human host [2]. These unique metabolic properties (the
human suffering. Despite significant advances in
pathways and the enzymes) may be exploited for therapeutic
understanding the disease and the parasite, malaria still
intervention and new antimalarial drug discovery. The
remains one of the leading causes of morbidity and
malaria parasite mainly grows and proliferates within the
mortality, particularly in the tropics with a staggering 500
host erythrocytes. This is commonly referred to as
million new infections and 1 million deaths annually.
“intraerythrocytic schizogony” and is primarily responsible
Almost half of the world’s population is currently at the risk
for pathogenesis of the disease [3]. Clinical manifestations
for malaria infection. Although malaria incidence is mostly
of malaria result from continuous proliferation of the parasite
centered in tropical regions, the impacts, especially
within the host erythrocytes and with rupture of schizonts
economic, of the disease are global. Recent trends indicate
[3]. The drugs acting on this part of parasite life cycle and
rapid emergence of drug-resistant and more virulent strains
clearance of the parasite from the blood are therefore referred
of the parasite to further intensify the problem [1]. The
to as blood schizontocidal antimalarials [4]. Almost all
choice of therapies currently available for the treatment of
antimalarials currently in use belong to this class; an
malaria is highly limited, and several of these may
exception is the 8-aminoquinoline class which act on
eventually be lost or compromised due to drug resistance.
exoerythrocytic stages of the parasite and are used for
An armament of new antimalarial drugs with proven clinical
treatment of relapsing malaria [5]. During intraerythrocytic
efficacy against current drug-resistant cases of malaria
phase of the malaria life cycle hemoglobin is utilized as a
including Plasmodium falciparum as well as Plasmodium
predominant source of nutrients. The amino acids derived
vivax infections is critical to combat the disease and cope
from digestion of hemoglobin are incorporated into parasite
with the problem of further development of resistance.
proteins and may also be utilized for energy metabolism.
The unique life cycle of the malaria parasite and the Massive degradation of about 5mM hemoglobin releases
resulting microenvironments within the host, vector and the large amount of toxic free heme [6]. Continuous degradation
of hemoglobin and concomitant detoxification of heme are
*Address correspondence to this author at the National Center for Natural absolutely necessary for uninterrupted growth and
Products Research, School of Pharmacy, University of Mississippi, proliferation of the parasite. Therefore, the metabolic
University, MS 38677, USA; Tel: 1-662-915-7882; Fax: -1-662-915-7062; functions related to hemoglobin digestion and heme
E-mail: btekwani@olemiss.edu

1386-2073/05 $50.00+.00 © 2005 Bentham Science Publishers Ltd.


64 Combinatorial Chemistry & High Throughput Screening, 2005, Vol. 8, No. 1 Tekwani and Walker

detoxification pathways may be potential targets for new the hemoglobin molecule is relaxed the plasmepsins (I, II)
antimalarial drug discovery [6]. Protease inhibitors, which and the falcipains can further cleave at other sites of the
can inhibit hemoglobin digestion, have shown promising protein. Four genes coding different falcipains have been
antimalarial properties in vitro as well as in vivo [7]. identified in P. falciparum genome. Falcipain 2 and
However, involvement of multiple proteases and gaps in the possibly falcipain 3 have been identified as a major
knowledge of the specific roles of each in hemoglobin trophozoite food vacuole cystein proteases [19]. Multiple
degradation, have limited their clinical applications. Broad orthologs of the falcipains have been identified in other
substrate specificity of the enzymes and poor selectivity of plasmodial species. The corresponding cysteine proteases
the inhibitors will need further study before successful identified in P. vivax have been termed as vivapain 2 and
development of protease inhibitors as antimalarials. vivapain 3 [20]. Falcipain 2 and falcipain 3 localize to the
Discovery of a unique heme detoxification process in malaria food vacuole, are active at acidic pH of the food vacuole, and
parasites, which involves incorporation of heme into a readily hydrolyze native and denatured hemoglobin [19]. P.
crystalline complex called hemozoin or the malarial falciparum contains two nearly identical copies of the
pigment, and identification of this pathway as a target for falcipain 2 gene. Falcipain 2 and 3 are 66.6% identical in
action of several known antimalarials have generated their catalytic domains, but they differ in stage specificity,
significant interest in this pathway for new antimalarial drug with maximal expression of falcipain 2 in early trophozoites
discovery [8]. A few recent review articles have highlighted and expression of falcipain 3 is more mature parasites [19].
the mechanism of formation of hemozoin, a crystalline A multispecificity protease substrate was used to profile the
pigment and the major product of heme detoxification proteolytic activities in P. falciparum in a stage dependent
pathway in malaria parasite [9-12]. However, the exact manner using a combination of fluorescence and MALDI
mechanism of formation of hemozoin within the parasite mass spectrometry [21]. Cysteine protease activity was
food vacuole, the physiological site of hemoglobin shown to be dominating at neutral pH, whereas aspartic
degradation and hemozoin synthesis, is still a subject on protease activity contributed predominantly to the
intense debate. This review focuses primarily on different proteolytic repertoire at acidic pH. Maximum proteolysis
experimental approaches used for the assay of hemozoin was observed at the trophozoite stage followed by the
formation in vitro. High throughput screening (HTS) schizonts and the rings.
approaches for in vitro hemozoin assays may be useful for It appears that the actions of plasmepsins and falcipains
identification of novel inhibitors of this parasite-specific within the digestive vacuoles, produce small peptides
pathway as potential antimalarial agents. instead of amino acids [22]. The resulting globin peptides
serve as the substrate for the zinc metalloprotease, falcilysin
2. HEMOGLOBIN DIGESTION BY MALARIA (FLN) [23]. FLN prefers to degrade small globin peptides up
PARASITE: ROLE OF MULTIPLE PROTEASES to 20 amino acids in length at pH optimum of 5.2,
consistent with the acidic environment of the food vacuole.
The malaria parasite depends on the host to fulfill its The temporal expression pattern of FLN is similar to that of
requirements of amino acids for synthesis of proteins and other globinases, yet it differs in the absence of proteolytic
other metabolic functions. Hemoglobin is the most abundant processing during biosynthesis. Random peptide substrate
protein in the erythrocytes cytoplasm (5 mM) and provides analysis has confirmed that FLN is highly active at acidic
the major source of amino acids [13]. The malaria parasite, pH, which is consistent with its role in hemoglobin
during intra-erythrocytic development and proliferation, degradation. However, the enzyme was found to be robustly
ingests more than 75% of the hosts hemoglobin. As an active at neutral pH, but with substantially different
estimate, at 20% parasitemia about 110 g of hemoglobin substrate specificity. FLN is localized to the parasite food
would be consumed by the parasite during 48 hours. The vacuole and is also associated with the vesicular structures
parasite ingests hemoglobin by the process of pinocytosis elsewhere within the parasite. It has been proposed that FLN
via the cytostome [14]. Ingested hemoglobin is degraded may have expanded role beyond globin catabolism and may
inside the acidic digestive vacuoles through a specific semi- function as two different proteases in its two locations [24].
ordered sequential process, which involves multiple Further hydrolysis of the digested hemoglobin by FLN
proteases [15]. Digestive vacuoles in the parasite are acidic action still generates small peptides, not the free amino acids
organelles with a pH estimated at 5.0-5.4 [16]. Different for utilization by the parasite. As no exopeptidase activity
aspartic proteases (plasmepsins), cysteine proteases has been detected in the parasite digestive vacuole, it has
(falcipains) and a metalloprotease (falsilysin) have been been assumed that these peptides translocate in parasite
characterized from the digestive vacuoles of P. falciparum cytosol where cytosolic neutral exo-peptidases complete the
[17]. Both plasmepsin I and plasmesin II are able to cleave digestion of peptides to individual amino acids for their
native non-denatured hemoglobin [18]. The degradation of subsequent utilization for parasite growth and maturation
hemoglobin is initiated by plasmepsin I, which hydrolyses [22]. A neutral aminopeptidase with apparent molecular
the 33Phe-34Leu bond in the hinge region, that appears to mass of 80+10 kDa has been identified in several species of
be vital for integrity of the hemoglobin tetramer. Eight malaria parasite [25]. A gene encoding for an aminopeptidase
additional plasmepsin genes have been identified in the P. belonging to the M1 family of zinc-metallopeptidases has
falciparum genome. A histo-aspartate protease (HAP) and been identified in P. falciparum [26]. The deduced amino
plasmepsin IV are homologous to plasmepsin I and II. HAP acids sequence corresponds to a 1056 residue protein with a
and plasmepsin IV were also found to be active in P. predicted molecular mass of 122 kDa. The antisera raised
falciparum food vacuole and may be the additional aspartate against a peptide encoded by this gene labelled two schizont
proteases involved in hemoglobin digestion [18]. As soon as proteins with molecular mass of 96 and 68 kDa, which may
Hemozoin Synthesis Pathway for New Antimalarial Drug Discovery Combinatorial Chemistry & High Throughput Screening, 2005, Vol. 8, No. 1 65

be the processed forms of the aminopeptidase. The protein phagocytic capacity and also alter cytokine secretion
purified from extracts of P. falciparum schizont culture pathways. Hemozoin has been shown as a potent
displayed aminopeptidase activity. The mechanism of proinflammatory agent in vivo, which could contribute to the
translocation of small peptides to the parasite cytoplasm is immunopathology related to malaria [39]. The hemozoin is
still not clear. It has been proposed that Pfmdr 1, which has predominantly accumulated in liver and spleen of the host,
been localized to the food vacuole membrane and is also a where amount of hemozoin increases with the progress of
member of ATP-binding cassette transporter family, might parasitaemia [40]. The molecular structure and mechanism of
be involved in pumping out the peptides from the food hemozoin synthesis have been discussed in more details in
vacuoles into the cytoplasm [27]. the subsequent sections.
Massive degradation of hemoglobin by the parasite Other pathways proposed for detoxification involve non-
generates large amount of free heme as a toxic by-product. enzymatic [41, 42] as well as enzymatic [43] degradation of
As soon as heme is free from its protein scaffold it attains heme to non-toxic metabolites. Figure 1 illustrates different
toxic characteristics [28]. The biochemical consequences of pathways suggested for detoxification of heme by the
accumulation of non-sequestered heme include membrane malaria parasite. As an estimate only about one third of the
lysis and inhibition of parasitic proteases [29-31]. heme released due to hemoglobin degradation may be
sequestered as hemozoin. The remainder may be detoxified
by alternate pathways [41-43]. A possible route has been
3. HEME DETOXIFICATION PATHWAYS OF THE proposed through degradation of heme by a non-enzymatic
MALARIA PARASITE hydrogen peroxide mediated process (Fig. 1, pathway 2)
The intra-erythrocytic asexual stage of the malaria which may occur within the parasite food vacuole. When
parasite resides in environment rich in hemoglobin and is oxyhemoglobin is released into the food vacuole of the
exposed to toxic amounts of heme when hemoglobin parasite at pH 5.2, it is rapidly converted to methemoglobin.
undergoes degradation, as occurs spontaneously in hemolytic Release of an electron and oxygen as a result of this reaction
anemia with the production of Heinz bodies [32]. It was generate superoxide anion. The superoxide anion will
shown that concentrations of heme as low as 20 µM will dismutate to hydrogen peroxide at pH 5.0. The food
lyse malaria parasites within 10 minutes; this amount of vacuoles may be transiently protected from the oxidative
heme can be produced by the destruction of only 0.1% of stress by the antioxidant enzymes viz., superoxide dismutase
total hemoglobin present in erythrocytes [33]. The and glutathione reductase, acquired from the erythrocyte
trophozoite stage of malaria parasite consumes almost 75% cytoplasm. These enzymes may be rapidly degraded by
hemoglobin of erythrocyte, with concomitant release of free vacuolar proteases and the lipids and proteins within the
heme. With heme as a prosthetic group of hemoglobin, the food vacuoles would be susceptible to the oxidative damage.
iron is in the ferrous state, but once heme is free from its Free heme may react with hydrogene peroxide displaying
protein scaffold it tends to loose one electron and assumes both catalase and peroxidase-like activities that regenerate the
the ferric state [34]. This ferric heme could promote heme molecule. Alternatively, heme may also undergo
membrane damage via its peroxidative properties [35] and peroxidative reaction that causes destruction of porphyrin
may also interfere with the hemoglobin degradation ring. It has been shown that under the physiological
pathway. The cysteine protease, falcipain has been shown to conditions similar to that of the parasite food vacuole, free
be very sensitive to free heme [6, 36]. heme undergoes rapid peroxidative decomposition [41].
Another non-enzymatic mechanism proposed for degradation
To avoid the damaging properties of free heme it is and detoxification heme is through its glutathione mediated
necessary for the parasite to convert it to non toxic degradation [42]. Presence of heme oxygenase, which
metabolites. Several properties of hemozoin, which is converts heme to biliverdine, has been reported in the rodent
structurally similar to β-hematin, make it an ideal excretory species of malaria parasites (43), but no heme oxygenase has
product of heme detoxification. First it is dense and been found in P. falciparum. Glutathione-mediated and
insoluble under physiological conditions and this may be an enzymatic degradation have been proposed to occur out side
irreversible link between hemoglobin degradation and release the parasite food vacuole. However, no evidence has been
of heme. Due to the coordinated nature of heme in, β- given for translocation of the heme across the food vacuoles.
hematin it does not contribute to oxygen radical stress and Part of the free heme has been found bound to the parasite
auto-oxidation. membranes. Possibly, both hemozoin synthesis as well as
Detoxification of heme occurs through different heme degradative pathways operate simultaneously in the
pathways. The most important and the predominant malaria parasite and are inhibited by the antimalarials known
mechanism is sequestration of heme into “hemozoin”, also to interfere with heme detoxification functions of the
known as the malaria pigment. Heme is incorporated into a parasite. Egan et al. [44] have shown that 92% of the iron
crystalline black brown pigment which is accumulated present in P. falciparum infected erythrocytes was located
within the parasite food vacuole. Hemozoin may be within the parasite food vacuole and 88% of this was in the
visualized as regular fibrillar crystals by electron microscopy form of hemozoin. 57Fe-Mossbauer spectroscopy shows that
[37]. Appearance of hemozoin crystals may be used as the hemozoin was the only detectable iron species in
evidence for presence of the malaria parasite within blood trophozoites, these findings were further confirmed by
and may have importance in diagnosis of the disease [38]. electronic microscopic imaging [44]. Hemozoin synthesis
After schizont maturation when the erythrocyte ruptures, pathway may be of greater interest for new antimalarial drug
hemozoin is released into the circulation. It is initially discovery due to its uniqueness.
ingested by macrophages, where it appears to diminish
66 Combinatorial Chemistry & High Throughput Screening, 2005, Vol. 8, No. 1 Tekwani and Walker

Fig. (1). Different heme detoxification pathways suggested in malaria parasite. Pathway (1) Biocrystallization of heme to hemozoin,
an insoluble black brown pigment, is the most predominant mechanism of heme detoxification [44]; Pathway (2) Peroxidative
degradation of heme [41]- Under acidic conditions of food vacuoles FP-Fe(II), released from hemoglobine is instantly oxidized to Fe-
(III). Transfer of electron to oxygen generates a superoxide radical, which is spontaneously converted to hydrogen peroxide.
Peroxidative cleavage reaction destroys the porphyrine ring; Pathway (3) Glutathione mediated heme degradation [42]; Pathway(4)
Enzymatic degradation of heme to bilirubin [43]. The later two mechanisms require translocation of free heme from digestive vacuole
to the cytosol, which is yet to be established.

4. MOLECULAR CHARACTERISTICS OF the structure of hemozoin. The IR spectrum of hemozoin


HEMOZOIN: AN ORDERED CRYSTALLINE was significantly different from that of hematin or heme and
STRUCTURE contained additional peaks at 1664 and 1210 cm-1, a
characteristic of Fe-carboxylate bond. Solubility properties
Hemozoin is not identical to hematin
of hemozoin are similar to a derivative of heme called β-
(Ferriprotoporphyrin IX hydroxide), as was speculated
hematin. Hemozoin and β-hematin show similar elemental
initially [45] since they show very distinct solubility
composition, infrared spectra, and also extended x-ray
properties. Hemozoin is insoluble in weakly basic
absorption fine structure spectra [46]. The x-ray powder
bicarbonate buffer and aprotic solvents such as dimethyl
diffraction pattern obtained from β-hematin was also
sulfoxide and pyridine [46]. In contrast hematin is readily
indistinguishable from that of hemozoin [46]. Laser Raman
solubilized in both types of solvents, either by solvation of
spectroscopy, which has been used as a powerful technique
carboxylate side chains or by solvent coordination with the
for elucidation of both structural and functional information
ferric iron. It was also proposed that hemozoin is composed
on heme and other metalloporphyrins [50], also showed that
of some parasite-derived proteins or peptides, which might
the spectrum of hemozoin is identical to the spectrum of β-
have a role in the sequestration of heme [47, 48]. However
hematin at all applied excitation wavelengths [51]. An
hemozoin is resistant to non-specific proteolysis and its
unexpected observation of dramatic band enhancement at the
elemental composition was found to be very similar to heme
excitation wavelength of 780 nm could be employed for
[46, 49]. Proteins associated with hemozoin might have
Raman imaging of hemozoin in the parasite food vacuole
bound to the pigment non-specifically during the process of
using intact P. falciparum infected erythrocytes. This
isolation. Hemozoin isolated from parasite extracts or tissues
technique enabled investigation of hemozoin within its
(liver and spleen) of malaria infected animals employing
natural environment. These observations indicate that
non-denaturing sucrose density gradient exhibit totally
hemozoin and β-hematin both are structurally similar and
different spectrum of proteins associated with it
posses an ordered crystalline structure. Based on solubility
(unpublished observations). Hemozoin isolated from the
characteristics, infra-red (IR) spectroscopy and x-ray
parasite lysate and that accumulated in the liver and spleen
absorption pattern, the crystalline structure of hemozoin or
has shown similar elemental composition, solubility
β-hematin was initially proposed as a polymer of heme [46,
properties and infrared spectroscopic characteristics [40].
49]. Heme (Ferriprotoporphyrin IX, FP IX) units (Fig. 2A)
Slater et al. [46] used infrared spectroscopy, x-ray diffraction
in β-hematin are linked to each other through a coordinate
absorption spectroscopy and chemical synthesis to determine
Hemozoin Synthesis Pathway for New Antimalarial Drug Discovery Combinatorial Chemistry & High Throughput Screening, 2005, Vol. 8, No. 1 67

Fig. (2). (A). Free ferriprotoporphyrin IX, (B). Reciprocal Fe-carboxylate ferriproporphyrin IX dimer, the basic units of β-hematin or
hemozoin, as per the structure suggested by Pagola et al. [53]. (The pictures provided by Dr Simon J. Hocart).

bond, between ferric iron of one unit to propionate carboxyl dimers are also linked with each other through hydrogen
group of another [46]. Further, in situ analysis of hemozoin bonds between remaining carboxylate groups [54] providing
in intact desiccated malarial trophozoites by synchronized x- the characteristic crystalline structure to the pigment (Fig.
ray powder diffraction supported the polymeric nature of the 3A, B). Analysis of phase homogeneity and crystal
malaria pigment [52]. This study showed that two chains of morphology of β-hematin confirms the ordered crystalline
hemozoin are also associated by intermolecular hydrogen structure of the pigment [55].
bonding between uncoordinated carboxyl side chains. A later Synthetic β-hematin formed by different chemical
study, which used simulated annealing technique to analyze methods has shown marked morphological differences when
the powder diffraction data, suggested that the crystals of β- analyzed by scanning electron microscopy [55]. The β-
hematin were constituted of cyclic heme dimers [53]. The hematin crystallites formed from the acid precipitation
heme units in these dimers are linked to each other through reaction have few distinguishable faces and have upper
reciprocal iron-carboxylate bonds (Fig. 2B) and different
68 Combinatorial Chemistry & High Throughput Screening, 2005, Vol. 8, No. 1 Tekwani and Walker

estimate of 0.2 µm for the largest visible crystal dimension. absorption spectroscopy [59]. However, these oligomeric
In contrast, the crystallites resulting from base-mediated aggregates are different from β-hematin and may be separated
reaction have maximum lengths about 20 fold larger. These from it by their differential solubility in NaHCO3/Na2CO3
crystallites have more ordered morphology and are of regular buffer solutions. The micellar aggregates of hematin dissolve
size throughout the samples than those formed under acid in this solution while β- hematin does not. In aqueous
precipitation reaction. The pigment isolated from the human solutions conversion of heme to β-hematin may be
malaria parasites viz., P. falciparum, P. vivax, P. ovele and chemically achieved only under supraphysiological
P. malariae, the primate malaria parasite P. knowlesi and conditions. Chemical synthesis of β-hematin may be
the rodent parasite P. yoelii have a regular, flate-faced achieved under aqueous as well as non-aqueous conditions.
cuboidal morphology with modest size differences as Incubation of hematin in glacial acetic acid at 70-80OC for
analyzed by scanning electron microscopy [56]. However, about 18 hours yields formation of β-hematin. About 40-
the hemozoin crystals isolated from the avian malaria 50% of hematin is polymerized into β- hematin by this
parasite P. gellinaceum exhibit regularly irregular barrel process. The process may be termed as the thermal
shape with a waffle surface. Homozoin isolated from dehydration [59]. β-Hematin has also been prepared
different malaria parasite species also shows variable chemically in high yield by abstraction of HCl from hemin
susceptibility to degradation by hydrogen peroxide. These with a non-coordinating base in strictly anhydrous
morphological differences have been attributed to the conditions [59]. Egan et al. [60] proposed formation of β-
different biochemical environments within the parasites. The hematin in malaria parasite as a spontaneous chemical
different shapes and sizes of hemozoin crystals in different reaction. They proposed that formation of β-hematin can
malaria parasites may distinguish diverse malaria species occur spontaneously between 6 and 65 OC in 0.1-4.5M
[56]. acetate and pH 4.2-5.0. They also demonstrated that in vitro
synthesis of β-hematin was essentially complete within 30
min at 600C, 2 hours at 37 OC, and 8 days at 6 OC in
5. MECHANISM OF HEMOZOIN FORMATION IN absence of any parasitic material, in 4.5 M acetate buffer of
THE MALARIA PARASITE: PERSISTING DEBATE pH 4.8. In low acetate buffer (0.5M) the rate of reaction is
This is the most important and intriguing aspect related slow and it is 50% complete after 7 days of incubation at 37
OC. In buffers of higher ionic strength the concentration of
to hemozoin synthesis pathway, but is still under extensive
debate. Several theories have been proposed to explain the solubilized hematin is greater and this may be the reason of
mechanism of hemozoin formation by the malaria parasite. higher reaction rate in high acetate medium. This reaction
The concept of mechanism of hemozoin synthesis has been was also reported to proceed in a mixture of 0.1M glutamate
shifting along with the more accurate understanding of the and 0.4 M glycine, pH 4.5. By mossbaur spectroscopy, this
structure of the hemozoin. The very early idea of formation reaction was determined as zero order, instead of
of the malaria pigment was simply the association & autocatalytic [61]. Incorporation of β-hematin, in reaction
sequestration of free heme, released as a result of mixture does not show any effect on the amount of
hemoglobin degradation, with a protein (either a partially hemozoin formation. The products formed under above
digested peptide derived from hemoglobin or a parasite conditions were characterized by IR spectroscopy, but were
derived protein) [47]. Later, when hemozoin was not washed with the solvents, such as bicarbonate buffer and
characterized as a heme polymer [46], the proposal was made 2.5% SDS or DMSO, which distinguish heme aggregates
of an enzymatic reaction termed as ‘heme polymerization’ and β-hematin. However, formation of β-hematin does not
and a parasite derived proteinaceous factor named “heme occur spontaneously under physiological conditions, similar
polymerase” was proposed to catalyze this reaction [57, 58]. to those prevailing within the malarial digestive vacuoles
Formation of a product similar to hemozoin could be [62]. Later it was reported that, β-hematin can be
demonstrated in the presence of various factors related and spontaneously synthesized, if monomeric hematin is allowed
unrelated to malaria parasites and based on these reports to incubate for several days at 37 OC at pH-4.8, without any
different mechanisms of formation of malaria pigment have parasitic material. This product was characterized by washing
been proposed. The reports demonstrating in vitro synthesis with bicarbonate buffer and by IR spectra [63]. Still, these
of β-hematin may be mainly categorized into four groups studies could not explain that how synthesis of a material
viz., chemical synthesis and spontaneous formation of β- identical to β-hematin occurs so efficiently in the malaria
hematin; role of different proteins in initiation of β-hematin parasite under mild physiological conditions. The parasite
formation; formation of hemozoin by autocatalysis and digestive vacuole, the physiological site for hemozoin
finally initiation of heme polymerisation by different lipids. synthesis, has acidic pH, along with presence of organic
acids, lipids and proteins. In vitro formation of β-hematin
under aqueous acidic conditions and physiological
5.1. Chemical Formation of β-Hematin: A Non-
temperature may be achieved only in the presence of some
Physiological Process
biological factors viz., the malaria parasite lysate, organic
High resolution X-ray crystallographic studies and other solvent extracts of the parasite lysate, histidine rich protein
analysis have conclusively proved that hemozoin (the II or III, preformed hemozoin or β-hematin and also some
pigment isolated from the malaria parasite lysate) and β- unsaturated lipids [9, 10, 63-66]. These in vitro methods of
hematin (the synthetic pigment) are identical [46, 53]. This β-hematin formation, which simulate physiological
has facilitated the investigations on understanding the environment of the parasite digestive vacuole, along with
mechanism of pigment formation. The monomeric heme other cell biological studies have provided useful tools for
tends to aggregate in acidic solutions (pH<4.5) as gaining insight into this unique function of malaria parasite
determined by ultra-centrifugation, 1H NMR, and electron
Hemozoin Synthesis Pathway for New Antimalarial Drug Discovery Combinatorial Chemistry & High Throughput Screening, 2005, Vol. 8, No. 1 69

and understanding the physiological mechanism of result of this process. Accumulation of oxidative products of
hemozoin biogenesis. polyunsaturated lipids further supports this hypothesis [9].
5.2. Lipids Catalyzed β-Hematin Formation: A 5.3. Histidine Rich Proteins : The Nucleation Sites for β-
Physiochemical Oxidative Mechanism Hematin Formation
Hemozoin biosynthesis within the digestive vacuoles of A unique class of malarial histidine rich proteins (HRP)
the malaria parasite may be a lipid-catalyzed physiochemical with characteristic multiple tripeptide (ala-his-his) repeats
reaction. Fractionation studies suggest that lipid is the only have been found to be involved in heme detoxification
active substance in malaria erythrocyte which may catalyze function of malaria parasite [64]. Screening of a P.
β-hematin in vitro. All the factors promoting β-hematin are falciparum lambda gt11 cDNA expression library with the
recovered in the chloroform extract of P. berghei infected antiserum against hemozoin, identified the clones of HRP II
erythrocytes and none is in the remaining dilapidated which was subsequently found to initiate formation of β-
hemozoin or protein fractions [9]. The factors catalyzing β- hemetin crystals in vitro at acidic pH [64]. HRP III, the
hematin formation are also recovered into the acetonitrile analog of HRPII, also catalyzes similar reaction. The HRP II
extract of the P. falciparum [67]. The acetonitrile extract and HRP III consist of repetitive hexa-peptide sequences
catalyzes β-hematin formation in the absence of any pre- containing histidine and alanine pairs. Ala-His-His-Ala-Ala-
formed hemozoin. The acetonitrile extract from synthetic β- Asp, is the major repeat hexa-peptide in Pf HRP II, which
hematin prepared from normal erythrocytes extracts, appears 30 times. Pf HRP II also contains 18 repeats of tri-
trophozoite lysate and the purified malarial hemozoin also peptide, Ala-His-His. HRP III contains two distinct regions
promote β-hematin formation in vitro [65, 68]. The factors of tandem repeats separated by a gap of 26 amino acids. The
catalyzing β-hematin formation in acetonitrile extract of P. first region contains three repeats of Ala-His-His, two of
falciparum have been identified as methyl esters of oleic, Ala-His- His-Val-Ala-Asp and 13 repeats of Ala-His-His-
palmitic and stearic acids. The acetonitrile extract of normal Ala-Ala-Asn and one Ala-His-His-Ala-Ala-Asp. Second
erythrocytes also promotes β-hemetin formation [68]. The repeat unit in Pf HRP III is Asp-(Asp/Gly)-Ala-His-His,
phospholipids viz., phosphatidyinositol, phosphatidyl which is different from Pf HRP II. Both proteins contain a
serine, sphingomyelin and phospatidyl choline also catalyze hydrophobic leader peptide, which may have some role in
β-hematin formation [63]. The commercially available translocation of these proteins [71]. The hexa-peptide repeat
unsaturated fatty acids viz., arachidonic acid, linoleic acid, has been identified as the heme binding motif in HRPII
oleic acid and palmitoleic acids, and 1-mono and [72]. However linear monomers, dimmers and trimers of the
dioleoylglycerol are able to promote β-hematin formation [9, hexapiptide (Ala-His-His-Ala-Ala-Asp) failed to promote β-
66]. Tri-oleoylglycerol, cholesterol, dioleoyl hematin formation. They rather inhibited the reaction,
phosphatidylethanolamine and saturated fatty acids viz., probably through competing for heme [72]. In an effort to
stearic acid and palmitic acid do not promote this reaction. understand the mechanism of β-hematin formation by HRP
The unsaturated fatty acids and their mono- and diglycerides II it was demonstrated that some peptide dendrimers,
serve to concentrate monomeric heme and keep it in a designed on the basis of tandem tripeptide (ala-his-his)
favorable state for dimerization [9]. In vitro β-hematin repeat motif of PfHRPII, were able to bind heme and also
formation promoted by Plasmodium yoelii homogenate, the promoted β-hematin formation in vitro. Tandem
lipid extracts, and linoleic acid were blocked by ascorbic organization of tripeptide, the heme binding site, may be
acid, reduced glutathione, sodium dithionite, beta- necessary for initiating formation of β-hematin by PfHRP-II
mercaptoethanol, dithiothreitol, and superoxide dismutase [73]. Presence of HRPII in P. falciparum digestive vacuole
[69]. Oxidized glutathione did not show any effect. has been demonstrated by con-focal microscopy [64].
Preoxidized preparations of the lipids extracts or the P. However, major amount of HRP II produced by the malaria
yoelii homogenate failed to catalyze β-hematin formation. parasite is secreted out of the parasite and even out of the
Depletion of oxygen in the reaction mixtures also inhibited infected erythrocytes [74]. Immunochemical detection of
the lipid-catalyzed β-hematin formation. β-Hematin HRP II in plasma or serum of the infected patients is used in
formation was also inhibited by p-aminophenol, a free diagnosis of malaria [75]. It was assumed that HRP II is
radical chain reaction breaker. An oxidative mechanism has internalized inside the digestive vacuole with the
been proposed for lipid-mediated β-hematin formation, hemoglobin ingestion, where it binds with the heme released
which may be mediated by generation of some free radical after hemoglobin degradation and converts it into hemozoin.
intermediates of heme [69]. These reports suggest that the In another immunolocalization studies HRP II was mainly
factors responsible for hemozoin formation are not specific localized in the cytosol of infected erythrocytes. However a
to malaria parasite. Leinoleic acid or the derivatives of small sub-population of HRP II was co-localized with a food
linoleate may be the principal lipids that catalyze β-hematin vacuole associated protein, p-glycoprotein homologue (Pgh-
formation within the digestive vacuoles as the parasite is 1) [76]. Both native as well as recombinant Pf HRP II
rich in linoleic acid which may be derived from inner promote in vitro synthesis of hemozoin, whereas many other
membranes of hemoglobin laden endocytic vesicles [9, 70]. proteins with heme binding characteristics, including human
Heme released as a result of hemoglobin degradation histidine rich glycoproteins or bovine serum albumin, do
immediately co-precipitate with the unsaturated lipids. This not convert heme to β-hematin [64]. It has been recently
process and the microenvironment within the parasite hypothesized that HRPII initiates formation of β-hematin as
digestive vacuoles or the endocytic vesicles produce the a probable nucleation site, and the further growth of
necessary physiochemical environment for dimerization of hemozoin crystal involves unsaturated lipids [77]. However,
heme and the unsaturated lipids undergo oxidation as a some researchers failed to observe a synergistic action of
HRPII and mono-oleoyl glycerol in formation of hemozoin
70 Combinatorial Chemistry & High Throughput Screening, 2005, Vol. 8, No. 1 Tekwani and Walker

[9]. Further investigations are needed to ascertain the role of crystal formation [64, 65], which may subsequently grow
HRP in hemozoin synthesis. further through an auto catalytic mechanism [65].
Intraerythrocytic schizogony of malaria parasite and
5.4. A Cytostomal Pathway of Hemozoin Biogenesis:
ingestion of hemoglobin exhibit a unique cell biological
Role of Inner Membrane of Endocytic Vesicles
characteristic. Malaria parasite ingests host cell cytoplasm
Hampelmann et al. [78] have recently proposed a unique through cytostome, a unique mouth like sub-cellular
cytostomal pathway of bio-crystallization of heme to form structure [79]. This process results into formation of unique
hemozoin. The pathways is based on the perception that a double membrane transport vesicles, which ultimately fuse
nucleation center or template is essential for initiation of the with the parasite food vacuole and act as the carriers for the
transport of host derived nutrients, mainly hemoglobin [80].
This hypothesis considers that non-selective nutrient
permeable channels present on parasitophorous vacuolar
membrane (PVM) [81] generate an acidic environment
within the transport vesicles and digestion of hemoglobin is
initiated in these vesicles, even before they are fused with
the parasite digestive vacuole [79]. The lipids and proteins
present in PVM, which is the inner transport vesicle
membrane, provide the nucleation site for initiation of
hemozoin crystal formation. The electron microscopic
observation that hemozoin crystals present in side the
parasite digestive vacuoles are associated with membrane
fragments that are distinct from the food vacuole membrane
provided supporting evidence in favor of this hypothesis
[78]. Ultrastructural analysis of P. falciparum has shown
presence of hemozoin in digestive vacuoles as well as small
vesicles of the parasite. An earlier observation by Orjih [82]
that normal erythrocyte ghosts treated with NaOH or HCl or
acetic acid, termed as activated membranes, convert 70% of
heme to β-hematin in a cell free reaction system, may also
be presented in support of this hypothesis. The cytostomal
pathway involving lipids-catalyzed mechanism presents a
convincing hypothesis for biogenesis of hemozoin. Release
of unsaturated lipids from the inners membranes of
hemoglobin laden transport vesicles results in co-
precipitation of heme and its dimerization. Formation of
hemozoin in axenically grown malaria parasite trophozoites,
without erythrocytic membranes poses a question against
this hypothesis [83]. The medium used in axenic cultivation
of malaria parasite contains erythrocyte sonicate. The
erythrocyte membranes ingested from the medium may
provide the template and also generate the lipids necessary
for hemozoin formation. More conclusive experimental
evidences would be required to convince formation of
hemozoin through this interesting and unique mechanism.

5.5. Biomineralization or Biocrystallization Mechanism


Soon after rediscovery of structure of hemozoin as a
heme dimer [53, 55], and confirmation of hemozoin
formation as a non-enzymatic but still a biocatalytic reaction
[10, 12, 62], the process of hemozoin formation in malaria
parasite was redefined as biomineralization [84] or
biocrystallinzation [85]. The crystalline structure of
hemozoin (heme dimers crystallized through intermolecular
Fig. (3). (A). Hemozoin, (malaria pigment) is a crystalline
hydrogen bonding) (Fig. 3) may be compared with the ice
macromolecule consisting of repeating Fe-O reciprocal dimers
and sugar crystals. Hemozoin was therefore also termed as a
linked through van der Waals and hydrogen bonding
bio-crystal [12]. Egan et al. in a controlled study on
interactions [53, 54]. (B). A space filling representation of the
mechanism of β-hematin formation determined that the
hemozoin crystal made in SYBYL from the unit cell of
process occurs via rapid precipitation of amorphous hematin,
hemozoin [53], colored for depth to illustrate the grooves on
followed by its slow conversion to the crystalline β-hematin.
the growing surface where the interaction are thought to occur
They also showed that formation of iron-carboxylate bond
between aminoquinolines and the crystal. (The pictures
(generation of heme dimers) and formation of crystalline β-
provided by Dr Simon J. Hocart).
hematin through intermolecular hydrogen bonding, are
Hemozoin Synthesis Pathway for New Antimalarial Drug Discovery Combinatorial Chemistry & High Throughput Screening, 2005, Vol. 8, No. 1 71

essentially simultaneous [84]. These authors concluded that formation. The major variation in hemozoin formation
formation of β-hematin parallels many biomineralization assays performed by various labs has been at the level of use
processes and also suggested that hemozoin synthesis in of different reaction cocktails including the different catalytic
malaria parasite may also be a unique biomineralization factors in the assay and also at the level of processing of
process. Biomineralization describes the deposition of assay mixtures at the end of the reaction for quantification of
minerals within or outside the cells of living organisms β-hematin. Spectrophotometric [88-94], radioisotopic [95],
[86]. Biomineralization is the process by which fluorometric [96] and high performance liquid
microorganisms mediate and catalyze inorganic reactions so chromatography (HPLC)–based [97] assays have been
as to form new mineral assemblages. The goal of described for quantification of β-hematin. Monomeric heme
biomineralization as a remediation tool is to control and hemozoin/β-hematin may also be differentiated by
microbial processes and their environment so as to sequester fourier transformed infra-red (FT-IR) spectroscopy [46]. Use
and immobilize specific heavy metals within exceptionally of quantitative FT-IR has been also shown in determination
strong bonds of neo-formed mineral assemblages. In this of β-hematin [60]. Formation of hemozoin occurs inside the
way, the bound metals are no longer mobile within a digestive vacuole of malarial trophozoite which has an acidic
geological time-scale. Formation of β-hematin though fits environment. Heme tends to aggregate under acidic pH. The
well in this definition but biomineralization refers mainly to aggregates of heme can be separated from β-hematin by their
the formation of inorganic salts under controlled biological differential solubility in alkaline bicarbonate solutions [46].
conditions. Definitive role of proteins and/or lipids has been The miceller aggregates of heme dissolve in this solution
suggested in hemozoin biosynthesis and the process seem to while β-hematin does not. For assay of β-hematin formation,
follow at least two step mechanism i.e initiation or these differential physical and chemical characteristics of
nucleation of hemozoin formation followed by growth of the monomeric heme/heme aggregates and β-hematin may be
biocrystal. Biocrystallization may be suggested as more utilized. Most of the assays require separation of monomeric
appropriate term to describe the hemozoin formation in heme and β-hematin for quantification. Estimation and
malaria [78]. Biocrystallization, like any other quantification of hemozoin or β-hematin in presence of
crystallizations, is a process involving the steps of monomeric heme or the heme aggregates has also been
nucleation and growth, where molecules have to be brought described recently [98]. Different protocols employed for β-
into a supersaturated, dynamically unstable state [87]. hematin formation assays and their applicability for routine
Nucleation is a prerequisite and the first step to crystal and high throughput screening have been discussed.
growth, yet excess nucleation causes the production of a
large number of small crystals instead of a smaller number of
6.1. Necessary Physicochemical Conditions for In Vitro
useful ones. Variation in the size and shape of hemozoin
β-Hematin Formation Assays
crystals isolated from different species of malaria parasite has
been attributed to variable intracellular environment [56]. Formation of β-hematin in vitro may be achieved in
several ways. The in vitro β-hematin formation assays are
Significant advancements have been in determining
founded on a basic protocol which involves incubation of
chemical and molecular structure of the malaria pigments.
monomeric heme (hemin HCl or hematin) in an appropriate
Similarly, considerable knowledge has been generated with
buffer at acidic pH (pH 3.0 -5.5). Formation of β-hematin is
regard to determination of optimal physical, chemical and
initiated by addition of an appropriate nucleation or a
biochemical conditions and the microenvironment necessary
catalytic factor. The test compounds are also added to the
for formation of β-hematin, particularly simulating the
reaction mixtures. After incubation for 12-24 hours the
biochemical environment of the physiological site of
reactions are processed in different ways and the resulting β-
hemozoin synthesis and accumulation. However, exact
hematin is quantified by different methods. Most of the
mechanism of hemozoin formation within the parasite
protocols use separation of the β-hematin pellet by
digestive vacuoles and pin pointed role of specific parasite
centrifugation or filtration and sequential washing of the
and/or host derived proteins and/or lipids in initiation and
pellet with water or Tris-HCl/SDS solution and alkaline
growth of hemozoin crystals are yet to be determined.
bicarbonate solution or DMSO. Dorn et. al., [63] have done
a systematic comparison of different chemical and
6. EXPERIMENTAL APPROACHES FOR IN VITRO biochemical factors which nucleate or initiate or catalyze
β-HEMATIN FORMATION ASSAY: APPLICATION formation of β-hematin. The trophozoite lysate mediated
IN HIGH THROUGHPUT SCREENING reaction has been found unsuitable for β-hematin formation
inhibition assay due to presence of membranes, which may
Several experimental approaches have been describes for bind with the test compounds. Presence of preformed
determination of formation of β-hematin in vitro and hemozoin may also compromise with sensitivity of the
evaluation of different antimalarials as well as other test assay. The acetonitrile extract of parasite trophozoite lysate
compounds for inhibition of this reaction. The important has been found suitable for in vitro screening. Later, some
difference between monomeric heme and β-hematin is their commercially available unsaturated lipids particularly
differential solubility in organic and aprotic solvents and unsaturated fatty acids and mono-olyeol glycerol have also
also in SDS and mildly alkaline bicarbonate solutions [46, been found more suitable catalytic factors, than the parasite
49]. This property has been used for isolation and lipids extracts, for use in these assays [66]. The pH below
quantification of hemozoin from the malaria parasite lysate 5.5 is absolutely essential for formation of β-hematin.. Most
[46], tissues of malaria infected animals [40] and also in in of the assays use the solutions with low salt concentration
vitro β-hematin formation assays. Various groups have particularly 50-200 mM sodium acetate (pH 3.0 -5.5) and
employed different methods for the assay of β-hematin also use a suitable catalytic or nucleating factor for
72 Combinatorial Chemistry & High Throughput Screening, 2005, Vol. 8, No. 1 Tekwani and Walker

promoting β-hematin formation. The assays based on reaction mixture as 5-5.2. It is recommended to adhere to a
spontaneous formation of β-hematin, the biomineralization particular order of addition of the reagents as hemin-Cl,
tests, have also been described for in vitro screening and acetate buffer and finally the test compound solution. For
evaluation of the compounds as inhibitors of β-hematin estimation of β-hematin, after incubation for 18-24 h at
synthesis [ 89, 91, 92]. 37οC, all the contents of the master incubation plate are
transferred to a 96-well multiscreen filtration plate with the
use of a multi-channel pipette. The filtration plate is kept on
6.2. Spectrophotometric Assays a vacuum manifold and washed with 200 µl of DMSO. The
Different versions of spectrophotometric assay of β- β-hematin pellet retained on the filters is dissolved in 0.1N
hematin formation are available. All these assays are based NaOH. Finally, 150 µl from each well is transferred into a
on differential solubility characteristics of heme and β- fresh clear 96-well plate. The plates are read on a microplate
hematin. For spectrophotometric quantification of β-hematin reader at 405 nm for quantification of β-hematin. The
it is necessary to dissolve the pigment formed during the spectrophotometric assays described above do not use any
reaction. The only solvent, which can dissolve β-hematin, is catalytic or nucleation factor for promoting β-hematin
NaOH (0.1- 1N) solution. However, this results into formation. These assays are based on biomineralization
breaking of iron-carboxylate bond and conversion β-hematin mechanism of β-hematin formation [84]. Another version of
to heme. β-Hematin, therefore may be quantified filtration-based microplate assay has been described recently
spectrophotometrically as heme equivalents. Different assays by us [94]. The reaction conditions used in this assay are
vary at the level of use of different reaction cocktails. A low close to the physiological environment of the parasite
throughput assay based on the use of lysate of rodent malaria digestive vacuoles, where biogenesis of hemozoin really
parasite, P. yoelii, for promoting β-hematin formation has occurs. This assay uses lipids for promoting β-hematin
been described [88]. This assay involves use of formation. The physiological mechanisms suggesting the
centrifugation for pelleting the heme aggregates and β- role of lipids, particularly associated with the inner
hematin and washing of the pellet sequentially with Tris- membranes of the endocytic vesicles, have an edge over
HCl and Sodium bicarbonate/SDS solutions. This assay, other mechanism suggested for hemozoin biogenesis [10].
which is performed in test tubes with a total reaction volume Use of assay conditions, which are closer to physiological
of 1 ml, can not be performed in microplates and is not environment, are likely to provide better results. Initial
suitable for large scale or high throughput screening. Some validation of this assay indicates that sequential washing of
laboratories have also used spontaneous chemical formation the pellet or the retenate on the filters, which contains heme
of β-hematin, as described by Egan et al. [60]. The aggregates as well as β-hematin, with Tris/SDS and alkaline
spectrophotometric assay described by Basilico et al. [89] bicarbonate solutions is necessary for complete removal of
uses 96-well U bottomed microplates. Each well receives monomeric heme aggregates. No spontaneous formation of
200 µl of the assay mixture consisting of 400 nmol of heme β-hematin occurs when 100 µM of hemin-Cl (dissolved in
(dissolved in 0.1M NaOH), 50 µl of the test compound 0.1N NaOH) suspended in 100 mM acetate buffer (pH 5.0)
(compound:heme ratio between 1:1 to 1:8) and 50 µl of is incubated at 37οC for 15-20 h. Both, the pellet of the
acetic acid (0.8 mmol). Final pH of the reaction mix is 3.00 purified β-hematin and the pellet containing the mixture of
and plates are incubated at 37˚C for 24 h. The plates are then heme aggregates and β-hematin, show IR spectra
centrifuged at 3300 g for 15 min, the pellet is washed with characteristic of β-hematin. Pure preparations of heme and β-
DMSO and finally dissolved in 0.1M NaOH. The assay has hematin, when analyzed separately, may be differential by
been validated with standard antimalarials which are known FT-IR. But, IR spectroscopic analysis can not differentiate
to inhibit β−hematin formation. This assay uses non- between monomeric heme aggregates and β-hematin in a
physiological pH and is sensitive to chloride and phosphate mixture. Additional analysis by x-ray diffraction absorption
salts. The assay was further modified by Parapini et al. [92] spectroscopy and scanning electron microscopy may be
to overcome these short comings. At higher pH (above 6.0) important [55]. The filtration-based spectrophotometric assay
heme tends to exist predominantly in the form of µ-oxo is directly performed in Millipore Multiscreen® plates. This
dimers. This is likely to prevent formation of iron- reduces an additional step of transfer of the contents from the
carboxylate bond and formation of β-hematin. Under acidic reaction plate, as used in biomineralization assay [91].
pH this equilibrium is shifted to monomeric form, which Reaction mixture, in a total volume of 200 µl, contains
facilitates the formation of β-hematin. Use of hemin (heme- heme (100 µM), acetate buffer (100 mM, pH 5.0), and the
Cl) instead of hematin (heme-OH) which is dissolved in test compounds dissolved in DMSO. The assay may tolerate
DMSO in place of NaOH could overcome these problems. up to 5% DMSO. The reaction is initiated by addition of 1
Recovery of β-hematin is significantly increased by keeping µg of mono-oleoyl glycerol to each well. Use of mono-oleol
the pH of the reaction mixture at 5.00. However, even the glycerol provided more reproducible results than acetonitrile
modified assay [92] may not be suitable for high throughput extracts of the parasite lysate or other unsaturated fatty acids.
screening due to use of multiple centrifugation steps, which The controls without catalytic factor are also set-up. The
may not be compatible with the automation. The assay was plates are incubated at room temperature (24-28C) for 14
further modified as “ferriprotoporphyrine IX hours with constant shaking. At the end of incubation the
biomineralization” assay to make it suitable for application plate is put on the filtration manifold and vacuum is applied
to HTS [91]. The modified assay, which is performed in to stop the reaction and remove all the soluble contents.
flat-bottomed 96-well microplate, uses the reaction mixture This step also removes the colored compounds or the
containing 50 µl of 5 mg/ml hemin-Cl (freshly dissolved in extracts, which may interfere with spectrophotometric
DMSO), 100 µl of 0.5M Sodium acetate buffer (pH 4.4) and quantification of β-hematin. The plate is sequentially washed
50 µl of the test compound. This yields final pH of the with Tris-HCl/SDS solution and alkaline bicarbonate buffer
Hemozoin Synthesis Pathway for New Antimalarial Drug Discovery Combinatorial Chemistry & High Throughput Screening, 2005, Vol. 8, No. 1 73

pre warmed at 37oC. For dissolving final β-hematin pellet injected onto the HPLC column. Initial elution of
20 µl of 1N NaOH is added to each well and the plates are monomeric heme is obtained with the solvent containing 10
incubated at 37 C for 10 to 30 min. 180 µl Tris-HCl (100 mM TMAC, 10 mM HS, 40% CH3CN, 60% water and
mM, pH 7.4) containing 2.5 % SDS is added to each well. H3PO4 to adjust the pH to 2.5. The peak of monomeric
Dissolution of β-hematin pellet directly in 0.1N NaOH, as heme could be detected at 400 nm. Subsequent shift of pH
suggested earlier in radiometric assay [95] and also in of the mobile phase to 12.0 converts β-hematin to heme
spectrophotometric biomineralization assays [89, 91, 92], which may be eluted as another peak. Both remaining
may not dissolve the β-hematin pellet completely or would monomeric heme as well as β-hematin formed may be
require extended incubation or extensive vortexing. analyzed in this assay. HPLC assay shows linearity in the
Quantification of heme in NaOH/Tris-HCl/SDS solution range of 78 pmol to 20 nmol of heme. The HPLC assay
provides better sensitivity than in 0.1N NaOH alone [58]. though is highly sensitive and specific but is tedious and
The contents (150 µl) are transferred to a fresh flat bottomed therefore may not be used routinely for screening the
clear microplate and the plates are read at 405 nm on a compounds.
microplate reader for quantification of β-hematin as heme
equivalents. Spectrophotometric assays are robust,
6.5. Radioactive 14C-Heme Incorporation Assay
inexpensive and provide sensitivity almost comparable to
the tedious, cumbersome, more expensive radioisotopic and Initial experiments on understanding the mechanism of
HPLC based assays. These assays are also suitable for hemozoin synthesis by malaria parasite used incorporation of
screening colored and fluorescent compounds and also the 14C-labelled heme into β-hematin [57, 68, 63]. This assay
crude extracts, which may normally interfere with also is based on differential solubility characteristics of heme
quantification of heme. Several alternate approaches for in and β-hematin. A high throughput assay has been developed,
vitro β-hematin formation assays and quantification of β- which is based on incorporation of 14C-labelled heme into β-
hematin have also been discussed to provide comprehensive hematin. The assay uses 96-well filtration microplates and
information available of this subject and also to present a requires a Wallace 1450 MicroBeta liquid scintillation
comparative picture of different assays. counter. The semi-automated high throughput screening
assay uses the Zymark bioassay robot system and has been
adopted from an original heme polymerization assay [63].
6.3. Quantitative Infra-Red Spectroscopic Assay The reaction is carried out in 96-well microplates. Each well
Formation of β-hematin may be achieved spontaneously receives, in a total volume of 100 µl, 500 mM sodium
under non-physisological conditions (4.5 M sodium acetate acetate (pH 4.8), unlabelled heme (10 pmols), 0.56 nCi of
and 60oC) without any nucleating factor or catalytic agents 14C-hemin, acetonitrile extract of P. falciparum trophozoit
[60]. Monomeric heme and β-hematin may be differentiated lysate (10 µl) and the test compound added as DMSO
by their infrared spectroscopic characteristics [46, 60]. IR solution (10 µl). After overnight incubation at 370C
spectra of β-hematin show sharp bands at 1660 and 1207 contents of the plates are transferred into a MultiScreen DV
cm-1, which are absent in IR spectra of heme. Based on this filtration plate, filtered and the contents of each well are
property Egan et al. [60] have described a quantitative FT-IR washed once with 400 µl of the solution containing 100
assay for evaluation of antimalarial compounds in vitro on mM Sodium bicarbonate-0.2% SDS and twice with 200 µl
formation of β-hematin. The anti-malarial drugs quinine, of Tris-HCl (pH 7.5). Thorough washing of the pellet
chloroquine and amodiaquine were demonstrated to inhibit retained on the filers is necessary to remove non β-hematin
β-hematin formation in this assay. Due to non-physiological heme aggregates. Incorporation of 14C-heme into β-hematin
assay conditions used in this assay and also due to pellet retained on the filters may be monitored by
limitation of this assay for processing large number of scintillation counting. This is the only β−hematin formation
samples for IR analysis, the FT-IR assay may not be assay, which has been truly adopted for high throughput
applicable for routine large scale screening of the screening. The assay has been applied for screening of
compounds. However FT-IR analysis may be used to obtain synthetic compounds libraries and also microbial broths.
confirmatory results with the lead compounds selected from Some novel non-quinoline antimalarial pharmacophores have
the large scale screening. been identified through this assay. At the time of
development of this assay, the role of lipids in β-hematin
formation was still under investigation. Later studies have
6.4. HPLC Assay proved that lipids are the major components of acetonitrile
The HPLC method described for quantification of heme extracts of the parasite lysate [35], which are involved in
and β-hematin and also for β-hematin formation assay, is promoting β-hematin formation. Use of some commercially
also based on their differential solubility characteristics [97]. available unsaturated fatty acids has also been demonstrated
The assay involves ion-pair reverse chromatography, as the catalysts for promoting β-hematin formation in vitro.
utilizing tetramethyammonium chloride (TMAC) and It is therefore likely that this assay may be further simplified
heptane sulfonic acid (HS) as the ion-pair agents in the by use of these commercially available lipids in place of
presence of 40% acetonitrile, and is performed on a acetonitrile extracts of the P. falciparum lysate. The
polymeric-resin-based column with a phenyl bonded phase. radiometric assay is simple and sensitive and may also be
The reaction mixtures with appropriate concentration of used for screening the colored as well as fluorescent
heme are incubated in 167 mM sodium acetate (pH 5.0) and compounds. The radioactive assay however is still more
P. falciparum lysate. Reaction is terminated by addition of expensive (primarily due to high cost of 14C-heme) than
an equal volume of 10 mM TMAC/10 mM HS/40% spectrophotometric assay, involves risk of health hazards due
CH3CN/60% water/H3PO4 to pH 2.5 and 200 µl is directly
74 Combinatorial Chemistry & High Throughput Screening, 2005, Vol. 8, No. 1 Tekwani and Walker

to use of radioactivity and requires proper disposal of the discovered even before the discovery of the parasite [99].
radioactive material. Even though several questions, regarding exact mechanism
of hemozoin synthesis by the malaria parasite, still remain
6.6. Biocrystal Growth Assay
unanswered, convincing evidences have been produced on
The crystal growth assay developed by Sullivan and co- inhibition of heme detoxifications functions of the malaria
workers [98] is based on spectrophotometric determination parasite as a target for action of most of the blood
of growth of β-hematin crystal on pre-formed β-hematin seed schizontocidal antimalarials, which are currently in clinical
crystals. Synthetic β-hematin used as a seed in this assay is use [8, 11, 12]. Inhibition of heme detoxification function of
prepared by incubating heme in 4.5 M acetate solution at the malaria parasite would lead to accumulation of toxic
60oC as described earlier [60]. Preparation of a large single heme which would kill the parasite due to membrane lysis
batch of β-hematin has been recommended to minimize the and interference with other vital functions of the parasite [6,
variation in crystal growth due to quality of the seed. 35, 36]. Quinoline antimalarials have been found to be the
DMSO concentration in these assays is kept below 1%. This most consistent inhibitors of hemozoin synthesis. Several
may limit the use of the assay for testing the compounds, novel antimalarials also have been found to interfere with the
which are soluble in DMSO, at higher concentrations. The process of hemozoin formation and some inhibitors of
assay mixture contains 5 nmol of preformed β-hematin and hemozoin synthesis are being perused as novel lead
50 µM bovine hemin chloride, in 0.5 ml of 0.1M antimalarial structures [95] (Fig. 4).
ammonium acetate, pH 4.8 and incubated at 37oC for 12
Quinine, the oldest known antimalarial, led to the
hours. As in earlier assays, in this assay too quantification
discovery of chloroquine, the most widely used antimalarial
of β-hematin crystal growth is based on differential
drug almost for more than half a century. Chloroquine
solubility of free heme and crystalline β-hematin in sodium
though has lost its importance as an antimalarial due to
bicarbonate solutions. Another version of this assay [98] is
widely spread emergence of drug resistance [1]. Quinolines
based on differential spectrophotometric properties of heme
however are still considered as important lead structures and
and β-hematin crystals [58]. Large difference in absorbancy
several labs are working on synthesis of the analogs [100-
of soluble free heme vs. insoluble crystalline β-hemetin at
102]. Several theories have been proposed for the mechanism
400 nm allows quantification of free heme in the reaction
of antimalarial action of chloroquine and related
mixture without its separation from β-hematin. The reaction
antimalarials and also have been the focus of discussion of
is terminated by addition of concentrated sodium
some recent reviews [103-105]. Interaction of quinolines
bicarbonate/SDS solution and the reaction mixtures are
with heme has been extensively studied and well established
directly read at 400 nm. The absorbance of this mixture is
[106, 107]. Quinolines interact and form a non-covalent
directly proportional to quantity of remaining free heme,
complex with heme and also inhibit the synthesis of
which is readily soluble in SDS/bicarbonate solution, while
hemozoin. Chloroquine binds non covalently to the growing
β-hematin remains insoluble and does not interfere with
face of hemozoin crystal and prevents its further growth
determination of free heme. The reaction mixtures are again
[108, 109] (Fig. 5). A chloroquine induced masking of a
read at 400 nm after addition of 10N NaOH, which results
lipid, which promotes hemozoin formation, has also been
into decrystallization of β-hematin back to heme. For high
suggested as a mechanism of inhibition of hemozoin
throughput crystal growth inhibition test the assay may be
synthesis by this drug [110]. Other blood schizontocidal
performed in 24 well micro-plates with total reaction
antimalarial drugs viz., quinine, quinidine, halofantrine, and
mixture volume of 250 µl. The remaining monomeric heme
mefloquine also inhibit this process [90]. A significant
may be quantified after addition of 0.5 mL of concentrated
correlation has been established between antimalarial efficacy
sodium bicarbonate/SDS solution. This assay involves no
and inhibition of β-hematin formation by these
centrifugation and no changing of plates or tubes. The assay
antimalarials. Incubation of chloroquine with the purified
has been validated with several quinoline and non-quinoline
hemozoin, isolated from the tissues of malaria infected mice,
antimalarials which are known to act through inhibition of
also causes its dissolution [111]. Primaquine, an 8-
hemozoin synthesis and has been found to be suitable for
aminoquinoline, which primarily kills malaria parasite
high throughput screening. Performance of this assay with
growing in hepatic cells, does not inhibit β-hematin
use of other nucleating agents is yet to be determined. For
formation. However, some 8-aminoquinoline antimalarials
high throughput screening the assay may still require
including tafenoquine, which also show prominent blood
validation in 96 well microplates. This assay is unsuitable
schizontocidal action, inhibit this process [112].
for evaluation of test compounds which show significant
Bisquinoline analogs (1, 2) have been found to be the potent
absorbance at 400 nm. Sensitivity of this assay may be
inhibitors of β-hemetin formation and also active against
compromised due to use of preformed β-hematin. Quantity
both chloroquine resistant and susceptible strains of P.
of the substrate (heme) and β-hematin should be limited to
falciparum. Therefore, blood schizontocidal antimalarial
keep the final absorbance of the reaction mixture within the
action of quinolines may be attributed primarily to their
scale of the reader.
interaction with heme as well as hemozoin and subsequent
inhibition of hemozoin synthesis.
7. HEMOZOIN: THE ANTIMALARIAL DRUG Artemisinin, which was originally isolated from a
TARGET Chinese herb Artemisia annua, and its first generation
Synthesis of hemozoin by the malaria parasite as a non- derivatives viz., arteether, artemether and sodium artesunate
toxic metabolite of heme detoxification process is the most have provided useful alternatives for the treatment of drug
distinct characteristic of the parasite. Malaria pigment was resistant and complicated cases of malaria [113]. Artemisinin
has been shown to target different parasite functions.
Hemozoin Synthesis Pathway for New Antimalarial Drug Discovery Combinatorial Chemistry & High Throughput Screening, 2005, Vol. 8, No. 1 75

Cl
Cl

N
HN N
HN
N NH HN N

N (CH2)n
Ro 47-7737 (1) N
N
O Bis(4-(4-diethylamino-1-methylbutyl)aminoquinoline (2)

HO N
HN NH
O (CH2) 5 O
HO O OH H2 N NH2
Cl
OH OH Pentamidine (5)

HO
2,3,4,5,6-pentahydroxyxanthone (X5) (3) Clotrimazole (4)

OH

N N

N M N N M N
OH
N N
N HO2C HO
HO2C

Cyproheptadine (6) CO2H CO2 H


Metalloporphyrin IX (7) Metallodueteroporphyrin IX (8)
O
OH
O NC
HO OH
NC
O
HO
NC
O Axisonitrile-3 (11)
Diisocyanoadociane (10)
Ellagic acid (9) Cl
N N
O
HN
HO
N N N

O
Cl
OH Cl
Benzophenone (Ro 22-8014) (13)
Triarylcarbinol (Ro 06-9075 (12)
Fig. (4). Some novel inhibitors of malarial heme detoxification functions and antimalarial pharmacophores.

However, fast and selective antimalarial action of these drugs inhibition of hemoglobin digestion in the malaria parasite.
has been primarily attributed to the ferrous iron/heme However, artemisinin and some analogs failed to show
induced bioactivation of the enoperoxide bridge and inhibition of β-hematin formation in in vitro
formation of carbon-centered cytotoxic radicals. The reactive biomineralization assays [116]. The β-hematin formation
intermediates further cause alkylation of biomolecules such assays for evaluation artemisinin and analogs require
as heme and other target proteins or enzymes leading to additional washing of the β-hemetin pellet with ethanol (90).
death of the parasite. Uptake studies using 14C-labelled The role of heme iron in bioactivation of endoperoxide
artemisinin have shown that the drug taken up by the atimalarials and hemozoin as the primary target for their
malaria parasite is concentrated in hemozoin [114]. Covalent antimalarial action is still under debate.
binding of artemisinin with heme in hemozoin has also been Hydroxyxanthones have been identified as a novel class
suggested. Artemisinin has shown inhibition of β-hematin of antimalarial compounds [117]. A xanthone analog, 2, 3,
formation in vitro in some parasite lysate and lipids-based in 4, 5, 6, -pentahydroxyxantone (3) has shown antimalarial
vitro assays at high concentrations [115] and subsequent action against chloroquine-sensitive and multidrug-resistant
76 Combinatorial Chemistry & High Throughput Screening, 2005, Vol. 8, No. 1 Tekwani and Walker

strains of P. falciparum in vitro. A recent study has also affinity for binding with heme and also inhibits heme
demonstrated antimalarial activity of hydroxyxanthones in detoxification functions of malaria parasite [122].
vivo in P. berghei/mouse model [118]. The antimalarial Clotrimazole also enhances heme induced membrane damage
potency of hydroxyxanthones correlates with their ability to and inhibits β-hematin formation [123]. Some other
inhibit β-hematin formation in vitro. Hydroxyxanthones antifungal azoles viz., ketoconozole and miconazole, which
form soluble complexes with heme and prevent the show significantly lower antimalarial activity as compared to
precipitation of heme in aqueous solution at acidic pH of clotrimazole, also bind to heme with similar affinities.
5.2. A structure-activity relationship study has shown that However, a significant correlation was noticed between
carboxy-oxygen, the two aromatic systems, 4- and 5- inhibition of in vitro β-hematin formation and antimalarial
hydroxyl groups on these hydroxyxanthones are important activity of the antifugal azoles (Tekwani, B.L., unpublished
for their interaction with heme and inhibition of β-hematin observations). Potent antimalarial action of pentamidine (5),
formation [119]. a diamidine drug used for treatment of Pneumocystis carinii
pneumonia and leishmania infections, has also been
proposed due to its binding with heme and interference with
heme detoxification functions of the parasite [124].
Cyproheptadine (6), an antihistaminic antidepressant has
shown significant blood schizontocidal activity in vivo
against a lethal multidrug-resistant (MDR) strain of
Plasmodium yoelii nigeriensis. A combination of
cyproheptadine and chloroquine shows improved
antimalarial activity compared to treatment with either drug
alone. Cyproheptadine inhibits in vitro β-hematin formation
in a parasite lysate-based assay. Treatment of P. yoelli
infected mice with cyproheptadine also causes significant
decrease in hemozoin level in the infected erythrocytes [125].
The mechanism of the antimalarial action of cyproheptadine
and its enhanced antimalarial activity with chloroquine could
be due, in part, to their inhibitory effect on heme
detoxification function.
With the discovery of hemozoin as an antimalarial drug
target, a new class of compounds viz., metalloporphyrines
(7) and metallodueteroporphyrines (8) have emerged as
potential antimalarials. Marked elevation of erythrocytic zinc
protoporphyrine IX occurs in some of the anemias associated
with malaria disease protection. Zinc protoporphyrin IX,
binds heme and also inhibits seed β-hematin crystal
formation as well as crystal extension [125]. Gallium
protoporphyrin IX, gallinium deutereoporphyrine IX and tin
protoporphyrin IX show marked inhibition of β-hematin
formation in trophozite lysate based assay [126, 127]. These
compounds also show antimalarial activity in vitro in P.
falciparum cultures. Fe(II)-Protoporphyrine IX (FP-Fe II),
released as result of hemoglobin digestion, is immediately
oxidized and then incorporated into hemozoin (Fig. 1).
Fe(II)-Protoporphyrine IX has been identified as a novel
endogenous atimalarial which inhibits incorporation of heme
into β-hematin and also kills malaria parasite [128]. FP-Fe II
itself does not incorporate into β-hematin.
In vitro β-hematin formation assays have also been
applied to screening of natural products and also the extracts.
Ellagic Acid (9), an antimalarial compound isolated from
bark of Tristaniopsis calobuxus inhibits β-hematin
Fig. (5). The suggested mechanism of inhibition hemozoin formation. [129 ]. Screening of 178 plant extracts from the
crystal growth by quinoline antimalarials. The mode of binding pharmacopia of Bolivian ethnia Tacana through in vitro β-
or interaction between hemozoin and chloroquine (CQ) is hematin formation assay have identified some plant extracts
illustrated with CQ sitting on the growing face of hemozoin with significant inhibition of β-hematin formation and also
[108, 109].(The pictures provided by Dr Simon J. Hocart). promising antimalarial activity in vitro [130]. From a series
of antimalarial terpene isonitriles, isolated from marine
Clotrimazole (4), an imidazole containing antifungal sponges, the two potent compounds viz., diisocyanoadociane
drug, has also shown promising antimalarial activity in vitro (10) and axisonitrile-3 (11) have been found to interact with
[120]. The drug primarily acts by causing changes to the heme by forming a coordinated complex with FP iron and
calcium ion flux [121]. Clotrimazole has a relatively high also inhibit formation of β-hematin [131]. The two
Hemozoin Synthesis Pathway for New Antimalarial Drug Discovery Combinatorial Chemistry & High Throughput Screening, 2005, Vol. 8, No. 1 77

compounds also inhibit alternate peroxidative and should be helpful for making use of these assays for in vitro
glutathione mediated heme detoxification functions. screening and new antimalarial drug discovery research.
Screening of 42 compounds isolated from nine plants used
in South Africa for the treatment of malaria identified several
antimalarials that interfere with heme detoxification ACKNOWLEDGEMENTS
functions of malarial parasite. Most of the compounds were The new antimalarial drug discovery research in the
found to be isoquinolines, which is a novel class of author’s lab is supported by Center for Disease Control &
compounds acting through binding with heme [132]. Prevention, USA, under Cooperative Scientific agreements
High throughput screening of a library of more than 100, (U50/CCU418839 and UR3/CCU418652) and by United
000 compounds, which were randomly selected, through a States Department of Agriculture (USDA)-ARS, under
radiometric in vitro β-hematin formation assay have cooperative scientific agreement no 58-6408-20009. We are
identified some novel inhibitors. Follow up studies with the highly thankful to Dr Simon J. Hocart, Tulane University
lead compounds in in vitro P. falciparum cultures and also Health Sciences Center, New Orleans, for providing the
in the Plasmodium berghei murine model further identified pictures of heme, hemozoin and hemozoin-chloroquine
the hemozoin synthesis inhibitors with actual antimalarial structure models.
activity. A triarylcarbinol (Ro 06-9075) (12) and a related
benzophenone (Ro 22-8014) (13) that showed oral activity in REFERENCES
the murine model have been identified as novel antimalarial
pharmacophores [95]. [1] White, N.J. J. Clin. Invest., 2004, 113, 1084.
[2] Gardner, M.J.; Hall, N.; Fung, E.; White, O.; Berriman, M.;
Hyman, R.W.; Carlton, J.M.; Pain, A.; Nelson, K.E.; Bowman, S.;
Paulsen, I.T.; James, K.; Eisen, J.A.; Rutherford, K.; Salzberg,
CONCLUSIONS S.L.; Craig, A.; Kyes, S.; Chan, M.S.; Nene, V.; Shallom, S.J.; Suh,
B.; Peterson, J.; Angiuoli, S.; Pertea, M.; Allen, J.; Selengut, J.;
Discovery of unique metabolic functions and pathways in Haft, D.; Mather, M.W.; Vaidya, A.B.; Martin, D.M.; Fairlamb,
malaria parasite have provided valuable tools for molecular A.H.; Fraunholz, M.J.; Roos, D.S.; Ralph, S.A.; McFadden, G.I.;
target-based new antimalarial drug discovery research [2]. Cummings, L.M.; Subramanian, G.M.; Mungall, C.; Venter, J.C.;
Most of the antimalarials, which are currently in clinical use, Carucci, D.J., Hoffman, S.L.; Newbold, C.; Davis, R.W.; Fraser,
have been identified through random screening and empirical C.M.; Barrell, B. Science, 2002, 419, 498.
[3] Miller, L.H.; Good, M.F.; Milton, G. Science, 1994, 264, 1878.
approaches. Exact mechanism of selective antimalarial action [4] Warhurst, D.C., Parasitol. Today, 1995, 11, 204.
of these antimalarials is still obscure. However, most of the [5] Bhat, B.K.; Seth, M.; Bhaduri, A.P. Prog. Drug Res., 1984, 28,
antimalarials, which kill blood stages of malaria parasite, 197.
have shown a common property of interaction with heme [6] Francis, S.E.; Sullivan, D.J.; Goldberg, D.E. Annual Rev.
Microbiol., 1997, 51, 97.
and interference with heme detoxification functions of the [7] Rosenthal, P.J. Curr. Opin. Hematol., 2002, 9, 140.
malaria parasite. Despite of living in a pool of heme, which [8] Ziegler, J; Linck, R.; Wright, D.W. Cur. Medicinal Chem., 2001,
is generated as a result of hemoglobin digestion, the malaria 8, 171-189.
parasite depends on de novo biosynthesis of this important [9] Fitch, C.D. Life Sciences, 2004, 74, 1957.
co-factor [133]. The parasite therefore depends on some [10] Tripathi A.K.; Tekwani, B.L. J. Parasitic Dis., 1999, 23, 61.
[11] Egan, T.J. J. Inorg. Biochem., 2002, 91, 19.
unique heme detoxification mechanisms, to get rid of the [12] Sullivan, D.J. Jr. Biopolymers, 2002, 9, 129.
indigenous toxic heme pool. Hemozoin, the major product [13] Krugliak, M.; Zhang, J.; Ginsburg, H. Mol. Biochem. Parasitol.,
of the malarial heme detoxification pathway, is formed 2002, 119, 249.
possibly through a non-enzymatic, physiochemical [14] Slomianny, C.; Prensier, G.; Charet, P. Parasitology, 1985, 90, 579.
[15] Goldberg, D.E.; Slater, A.F.G.; Beavis, R.;Chait, B.; Cerami, A. J.
biocrytallization mechanism. Formation of hemozoin, which Exp. Med., 1991, 173, 961.
is identical to β-hematin (a synthetic analog), possibly [16] Ollioro, P.L.; Goldberg, D.E. Parasitol. Today, 1995, 11, 294.
requires a nucleation factor for initiation of the reaction and [17] Rosenthal, P.J. In Antimalarial chemotherapy-Mechanism of
follows an autocatalytic crystal growth. Availability of in action resistance and new directions in drug discovery, P.J.
vitro assays for quantitative determination of β-hematin Rosenthal, Ed. Human Press, Totowa, 2000, pp. 325-345
[18] Banerjee, R.; Liu, J.; Beatty, W.; Pelosof, L.; Klemba, M;
formation and use of these assays in in vitro screening has Goldberg, D.E. Proc. Natl. Acad. Sci. USA, 2002, 990.
been useful in identification of several novel antimalarial [19] Sijwali, P.; Rosenthal P.J. Proc. Natl. Acad. Sci. USA, 2004, 101,
pharmacophores. The Spectrophotometric assays, based on 4384.
quantification of β-hematin as heme equivalents, and the [20] Na, B.K.; Shenai, B.R.; Sijwali, P.S.; Choe, Y.; Pandey, K.C.;
Singh, A.; Craik, C.S.; Rosenthal, P.J. Biochem. J., 2004, 378, 529.
radioactive assays, which are based on incorporation of 14C- [21] Pattanaik, P.; Jain, B.;Ravindra, G.; Gopi, H.N., Pal, P.P., Balram,
heme into β-hematin, may be useful for high throughput H.; Balram, P. Biochem. Biophys. Res. Commun. 2003, 309, 974.
screening. Significant efforts have been made for [22] Kolakovich, K.A.; Gluzman, I.Y.; Duffin, K.I.; Goldberg, D.E.
standardization of biological and physiochemical conditions Mol. Biochem. Parasitol., 1997, 87, 123.
necessary for in vitro β-hematin formation assays. Synthesis [23] Kolakovich, K.; Duffin, K.L.; Goldberg, D.E. J. Biol. Chem.,
1999, 274, 32411.
and accumulation of hemozoin occurs inside the parasite [24] Murata, C.E.; Goldberg, D.E. J. Biol. Chem., 2003, 278, 38022.
digestive vacuoles. Therefore, the methods which use the [25] Curley, G.P.; O’Donovan, S.M.; McNally, J., Mullaly, M.,
assay conditions closer to the microenvironment within the O’Hara, H.; Troy, A.; O’Callaghan, S.A.; Dalton, J.P. J. Eukaryot.
parasite digestive vacuole, are likely to generate better Microbiol., 1994, 41, 119.
[26] Florent, I.; Derhy, Z.; Allary, M.; Monsingny, M., Mayer, R.;
results. The review on comprehensive and comparative Schrevel, J. Mol. Biochem. Parasitol., 1998, 97, 149.
accounts of different in vitro β-hematin formation assay [27] Volkman, S.K.; Cowman, A.F.; Wirth, D.F. Proc. Natl. Acad. Sci.
methods vis-à-vis advantages and limitations of each method USA, 1995, 92, 8921.
[28] Slater, A.F.G. Exp. Parasitol., 1992, 74, 362.
78 Combinatorial Chemistry & High Throughput Screening, 2005, Vol. 8, No. 1 Tekwani and Walker

[29] Meshnick, S.R. Ann. Trop. Med. Parasitol., 1996, 90, 367. [72] Pandey. A.V.; Joshi, R.; Tekwani, B.L.; Singh, R.L. Chauhan, V.S.
[30] Fitch, C.D. Trans. Am. Clin. Climatol. Assoc. 1998, 109, 97. Mol. Biochem. Parasitol., 90, 281.
[31] Orjih, A.U.; Chevli, R, ; Fitch, C.D. Am. J. Trop. Med. Hyg., 1985, [73] Ziegler, J.; Chang, R.T. Wright, D.W. J. Am. Chem. Soc., 1999,
34, 223. 121, 2395.
[32] Sills, M.R.; Zinkham, W.H. Arch. Pediatr. Adolesc. Med., 1994, [74] Howard, R.J.; Uni, S.; Aikawa, M.; Aley, S.B.; Leech, J.H.; Lew,
148, 306. A.M.; Wellems, T.E.; Rener, J.; Taylor, D.W. J. Cell Biol., 1986,
[33] Fitch, C.D., Ciba. Found. Symp., 1983, 94, 222. 103, 1269.
[34] Hargrove, M.S.; Whitaker, T.; Olson, J.S.; Vali, R.J.; Mathews, [75] Moody, A. Clin. Microbiol. Rev., 2002, 15, 66.
A.J. J. Biol. Chem., 1997, 272, 17385. [76] Papalexis, V.; Siomos, M.; Campanale, N.; Guo, G. Kocak, G.
[35] Stojiljkovic, I.; Evavold, B.D.; Kumar, V. Expert. Opin. Investig. Foley, M. Tilley, L. Mol. Biochem. Parasitol., 2001, 115, 77.
Drugs, 2001, 10, 309. [77] Pandey, A.V.; Babbarwal, V.K; Okoyeh, J.N.; Joshi, R.M.; Puri,
[36] Pandey, A.V.; Tekwani B.L.; Singh, R.L.; Chauhan, V.S. J. Biol. S.K.; Singh, R.L.; Chauhan, V.S. Biochem. Biophys. Res.
Chem. 1999, 274, 19383. Commun., 2003, 308, 736.
[37] Saliba, K.J.; Folb, P.I.; Smith, P.J. Biochem. Pharmacol., 1998, 56, [78] Hempelmann, E.; Motta, C.; Huges, R.; Ward, S.A.; Bray, P.G.
313. Trends Parasitol., 2003, 19, 23.
[38] Hanscheid, T.; Melo-Cristino, J.; Pinto, B.G. Am. J. Trop. Med. [79] Slomianny, C. Blood cells, 1990, 16, 369.
Hyg., 2001, 64, 290. [80] Yayon, A.; Timberg, R.; Friedman, S.; Ginsburg, H. J. Protozool.,
[39] Jaramillo, M.; Plante, I.; Ouellet, N.; Vandal, K.; Tessier, P.A.; 31, 367.
Olivier, M., J. Immunol., 2004, 172, 3101. [81] Desai, S.A.; Krogstad, D.J.; McCleskey, E.W. Nature, 1993, 362,
[40] Pandey, A.V.; Tekwani, B.L. ; Pandey, V.C., Biomed. Res., 1994, 643.
16, 115. [82] Orjih, A.U. Exp. Biol. Med., 2001, 226, 746.
[41] Loria, P.; Miller, S.;Foley, M.; Tilley, L. Biochem. J., 1999, 339, [83] Trager, W. Parasitol. Today, 2003, 19, 388.
363. [84] Egan, T.J.; Mavuso, W.W. Ncokazi, K.K. Biochem., 2001, 40,
[42] Ginsburg, H.; Famin, O.; Zhang, J.; Krugliak, M. Biochem. 204.
Pharmacol., 1998, 56, 1305. [85] Hampelmann, E.; Egan, T.J. Trends Parasitol., 2002, 18, 11.
[43] Srivastava, P.; Pandey, V.C. Int. J. Parasitol., 1995, 25, 1061. [86] Bauerlein, E. Angewandte Chemie Int Ed., 2003, 42, 614.
[44] Egan, T.J.; Combrinck, J.M.; Egan, J.; Hearne, G.R.; Marques, [87] Wolf, S.G.; Frenkiel, T.; Arad, T.;Finkel, S.E.;Kolter, R.; Minskey
H.M.; Ntenteni, S.; Sewell, B.T.; Smith, P.J.; Taylor, D.; van A. Nature, 1999, 400, 83.
Schalkwyk, D.A.; Walden, J.C. Biochem. J., 2002, 365, 343. [88] Pandey, A.V.; Singh, N.; Tekwani, B.L.; Puri, S.K.; Chauhan, V.S.
[45] Brown, W.H. J. Exp. Med., 1911, 13, 290. J. Pharm. Biomed. Anal., 1999, 20, 203.
[46] Slater, A.F.; Swiggard, W, J.; Orton, B.R.; Flitter, W.D.; Goldberg, [89] Basilico, N; Pagani, E.; Monti, D.; Olliaro, P.; Taramelli, D. J.
D.E.; Cerami, A.; Henderson, G.B. Proc. Natl. Acad. Sci. USA, Antimicrobial Chemther., 1998, 42, 55-60.
1991, 88, 325. [90] Tripathi, A.K.; Gupta, A.; Garg, S.K.; Tekwani, B.L. Life Sci.,
[47] Yamada, K.A.; Sherman, I.W. Exp. Parasitol., 1979, 48, 61. 2001, 69, 2725.
[48] Ashong, J.O.; Blench, I.P.; Warhurst, D.C. Trans Roy. Soc Trop. [91] Baelmans, R.; Deharo, E.; Munoz, V.; Sauvain, M.; Ginsbergh, H.
Med. Hyg., 1989, 83, 167. Exp. Parasitol., 2000, 96, 243.
[49] Fitch, C.D.; Kanjananggulpan, P. J. Biol. Chem., 1987, 262, 15552. [92] Parapini, S.; Basilico, N.; Pasini, E.; Egan, T.J.; Olliaro, P.;
[50] Spiro, T.G.; Li, X.Y. In Biological Applications of Raman Taramelli, D.; Monti, D. Exp. Parasitol., 2000, 96, 249.
Spectroscopy, T.G. Spiro, Ed; Wiley, New York, 1988; Vol. 3, pp. [93] Deharo, E.; Garcian, R.N.; Oporto, P.; Gimenez, A.; Sauvain, M.;
1-38. Jullian, V.; Ginsburg, H. Exp. Parasitol., 2002, 100, 252.
[51] Wood, B.R.; Langford, S.J.; Cooke, B.M., Glenister, F.K.; Lim, J.; [94] Tripathi, A.K.; Khan, S.I.; Walker, L.A.; Tekwani, B.L. Anal.
McNaughton, D. FEBS Lett., 2003, 554, 247. Biochem., 2004, 325, 85.
[52] Bohle, D.S.; Dinnebier, R.E.; Madsen, S.K. J. Biol. Chem., 1997, [95] Kurosawa, Y.; Dorn, A.; Kitsuji-Shirane, M.; Shimada, H.; Satoh,
272, 713. T.; Matile, H.; Hofheinz, W.; Masciadri, R.; Kansy, M.; Ridley,
[53] Pagola, S.; Stephens, P.W., Bohle, D.S., Kosar, A.D.; Madsen, R.G. Antimicrob. Agents Chemother., 2000, 44, 2638.
S.K. Nature, 2000, 404, 307. [96] Sullivan, A.D.; Meshnick, S.R. Parasitol. Today, 1996, 12, 161.
[54] Bohle, D.S.; Kosar, A.D.;Madsen, S.K. Biochem. Biophys. Res. [97] Berger, B.J.; Bendrat, K.; Cerami, A. Anal. Biochem., 1995, 231,
Commun., 2002, 294, 132. 151.
[55] Bohle, D.S.; Kosar, A.D.; Stephens, P.W. Acta Cryst., 2002, D58, [98] Chong, C.B.; Sullivan, D.J. Jr. Biochem. Pharmacol., 2003, 66,
1752. 2201-2212.
[56] Noland, G.S.; Briones, N.; Sullivan, D.J. Jr. Mol. Biochem. [99] Nye, E.R. J. Med. Biogr., 2002, 10, 81.
Parasitol., 2003, 130, 91. [100] Raynes, K. Int. J. Parasitol. 1999, 29, 367.
[57] Slater, A.F.G.; Cerami, A. Nature, 1992, 355, 167. [101] Foley, M. Tilley, L. Pharmacol. Ther. 1998, 79, 55.
[58] Chou, A.C.; Fitch, C.D. Life Sci., 1992, 51, 2073. [102] O'Neill, P.M.; Bray, P.G.; Hawley, S.R.; Ward, S.A.; Park, B.K.
[59] Bohle, D.S.; Helms, J.B. Bioche. Biophys. Res. Commun., 1993, Pharmacol. Ther., 1998, 77, 29.
193, 504. [103] Sullivan, D.J. Int. J. Parasitol., 2002, 32, 1645.
[60] Egan, T. J.; Ross, D.J; Adams, P.A. FEBS Lett., 1994, 352, 54. [104] Ginsburg, H.; Golenser, J. Redox Rep., 2003, 8, 278.
[61] Adams, P.A.; Egan, T.J.; Ross, D.C.; Silver, J.; Marsh, P.J. [105] Slater, A.F. Pharmacol. Ther., 1993, 57, 203.
Biochem. J., 1996, 318, 25. [106] Egan, T.J.; Ncokazi, K.K. J. Inorg. Biochem., 2004, 98, 144.
[62] Pandey, A.V.; Tekwani, B.L. FEBS Lett., 1996, 393, 189. [107] Vippagunta, S.R.; Dorn, A.; Matile, H.; Bhattacharjee, A.K.;
[63] Dorn, A, ; Vippagunta, S.R.; Matile, H., Bubendorf, A.; Karle, J.M.; Ellis, W.Y.; Ridley, R.G.; Vennerstrom, J.L. J. Med.
Vennerstom, J.L.; Ridley, R. Biochem. Pharmacol., 1998, 55, 737. Chem., 1999, 42, 4630.
[64] Sullivan, D.J.; Gluzman, I.V.; Goldberg, D.E. Science, 1996, 271, [108] Sullivan, D.J.; Gluzman, I.; Russel, I.Y.; Goldberg, D.E. Proc.
219. Natl. Acad. Sci. USA, 1996, 93, 11865.
[65] Dorn, A.; Stoffel, R.; Matile, H.; Bubendorf, A.; Ridley, R. Nature, [109] Buller, R.; Peterson, M.L.; Almarsson, O.; Leiserowitz, L. Cristal
1995, 374, 269. Growth Design, 2002, 2, 553-562.
[66] Fitch, C.D.; Cai, G.; Chen, F.; Shoemaker, J.D. Biochim. Biophys. [110] Fitch, C.D.; Chen, Y.; Cai, G. J. Biol. Chem., 2003, 278, 22596.
Acta, 1999, 1454, 31. [111] Pandey, A.V.; Tekwani, B.L. FEBS Lett., 1997, 402, 236.
[67] Bendrat, K.; Berger, B.J.; Cerami, A. Nature, 1995, 378, 138. [112] Vennerstrom, J.L.; Nuzum, E.O.; Miller, R.E.; Dorn, A.; Gerena,
[68] Ridley, R.G.; Dorn, A.; Matile, H.; Kansy, M. Nature, 1995, 378, L.; Dande, P.A.; Ellis, W.Y.; Ridley, R.G.; Milhous, W.K.
138. Antimicrob. Agents Chemother., 1999, 43, 598.
[69] Tripathi, A.K.; Garg, S.K.; Tekwani, B.L. Biochem. Biophys. Res. [113] O’Neil, P.M.; Posner, G. J. Med. Chem., 2004, 47, 2945.
Commun., 2002, 290, 595. [114] Hong, Y.L.; Yang, Y.G.; Meshnick, S.R. Mol. Biochem. Parasitol.,
[70] Fitch, C.D.; Cai, G; Shoemaker, J.D. Biochim. Biophys. Acta, 1994, 63, 121.
2000, 1535, 45-49. [115] Pandey, A.V.; Tekwani, B.L.; Singh, R.L.; Chauhan, V.S. J. Biol.
[71] Sharma, Y.D. Int. J. Biochem., 1988, 20, 471. Chem., 1999, 274, 19383.
Hemozoin Synthesis Pathway for New Antimalarial Drug Discovery Combinatorial Chemistry & High Throughput Screening, 2005, Vol. 8, No. 1 79

[116] Haynes, R.K.; Monti, D; Taramelli, D.; Basilico, N.; Parapini, S.; [125] Agrawal, R.; Tripathi, R.; Tekwani, B.L.; Jain, S.K.; Dutta, G.P.;
Olliaro, P. Antimicrob. Agents Chemother., 2003, 47, 1175. Shukla, O.P. Biochem. Pharmacol., 2002, 64, 1399.
[117] Ignatushchenko, M.V.; Winter, R.W.; Bachinger, H.P.; Hinrichs, [125] Iyer, J.K.; Shi, L.; Shankar, A.H. Sullivan, D.J. Mol. Med., 2003,
D.J.; Riscoe, M.K. FEBS Lett., 1997, 409, 67. 9, 175.
[118] Fotie, J.; Nkengfack, A.E.; Rukunga, G.; Tolo, F.; Peter, M.G.; [126] Martiney, J.A.; Cerami, A.; Slater, A.F. Mol. Med. 1996, 2, 236.
Heydenreich, M.; Fomum, Z.T. Ann. Trop. Med. Parasitol. 2003, [127] Begum, K.; Kim, H.S.; Kumar, V.; Stojilkovic, I.; Wataya, Y.
97, 683. Parasitol. Res., 2003, 90, 221.
[119] Kelly, J.X.; Winter, R.; Peyton, D.H.; Hinrichs, D.J. Riscoe, M. [128] Monti, D.; Vodopivec, B.; Basilico, N.; Olliaro, P.; Taramelli, D.
Antimicrob. Agents Chemother., 2002, 144-150. Biochemistry, 1999, 38, 8858.
[120] Tiffert, T.; Ginsburg, H.; Krugliak, M.; Elford, B.; Lew, V.L. [129] Dell’Agli, M.; Parapini, S.; Basilico, N.; Verotta, L; Taramelli, D.;
Proc. Natl. Acad. Sci. USA, 2000, 97, 331. Berry, C.; Bosisio, E. Planta Medica, 2003, 69, 162.
[121] Tiffert, T.; Staines, H.M.; Ellory, J.C.; Lew, V.L. J. Physiol., 2000, [130] Baelmans, R.; Deharo, E.; Bourdy, G.; Munoz, V.; Quenevo, C.;,
525, 125. Sauvain, M. Ginsburg, H. J. Ethonopharmacol., 2000, 72, 271.
[122] Huy, N.T.; Kamei, K.; Yamamoto, T.; Kondo, Y.; Kanaori, K.; [131] Wright, A.D.; Wang, H.; Gurrath, M.; Konig, G.M.; Kocak, G.;
Takano, R.; Tajima, K.; Hara, S. J. Biol. Chem., 2002, 277, 4152. Neumann, G.; Loria, P.; Foley, M.; Tilley, L. J. Med. Chem., 2001,
[123] Huy, N.T.; Takano, R.; Hara, S.; Kamei, K. Biol. Pharm. Bull., 44, 873.
2004, 27, 361. [132] Steele, J.C.P.; Phelps, R.J.; Simmonds, M.S.J.; Warhurst, D.C.;
[124] Stead, A.M.; Bray, P.G.; Edwards, I.G.; DeKoning, H.P.; Elford, Meyer, D.J. J. Antimicrob. Chemother., 2002, 50, 25-31.
B.C.; Stocks, P.A.; Ward, S.A. Mol. Pharmacol., 2001, 59, 1298. [133] Sato, S.; Clough, B.; Coates, L.; Wilson, R.J. Protist., 2004, 155,
117.

You might also like