ArticlePDF Available

Mitochondrial heterogeneity in human malarial parasite Plasmodium falciparum

Authors:

Abstract and Figures

Mitochondria of the human malarial parasite Plasmodium falciparum in sexual blood stages (or gametocytes) had been structurally different from those of asexual blood stages of their life cycle in human host. We report here the existence of mitochondrial heterogeneity based on their characteristics of ultrastructural morphology in the asexual and sexual blood stages of P. falciparum from in vitro continuous cultures. Mitochondria in the sexual stage-parasites were more numerous and contained a greater density of cristae than the organelles in the asexual stage-parasites. It was demonstrated that there were apparent variations in size and appearance of the mitochondria between the male and female parasites of the sexual gametocytic stages. Mitochondrial oxygen consumption of the sexual stage-parasites was relatively low, and it was not different from the asexual blood stage-parasites. However, both stages of the parasites' growth and their oxygen consumption were found to be sensitive to atovaquone, cyanide and 5-fluoroorotate which were inhibitors of mitochondrial electron transport system and pyrimidine biosynthetic pathway, respectively. Therefore, the role of mitochondrial organelles with different morphological properties in the asexual and sexual stages of parasite's development remains to be elucidated.
Content may be subject to copyright.
Mitochondrial Heter ogeneity in Human
Malarial Parasite
Plasmodium falciparum
Sudaratana R. Krungkrai a, Pr eecha Leangaramgul b, Sanya Kudan b, Phisit
Prapunwattana b and Jerapan Krungkrai b,*
aDepartment of Biochemistry, Faculty of Science, Rangsit University, Patumthani 12000, Thailand.
bDepartment of Biochemistry, Faculty of Medicine, Chulalongkorn University, Rama IV Road,
Bangkok 10330, Thailand.
* Corresponding author: E-mail: fmedijkk@md2.md.chula.ac.th
Received 11 Feb 1999
ABSTRACT Mitochondria of the human malarial parasite Plasmodium falciparum in sexual blood stages
(or gametocytes) had been structurally different from those of asexual blood stages of their life cycle in
human host. We report here the existence of mitochondrial heterogeneity based on their characteristics
of ultrastructural morphology in the asexual and sexual blood stages of P. falciparum from in vitro
continuous cultures. Mitochondria in the sexual stage-parasites were more numerous and contained a
greater density of cristae than the organelles in the asexual stage-parasites. It was demonstrated that
there were apparent variations in size and appearance of the mitochondria between the male and female
parasites of the sexual gametocytic stages. Mitochondrial oxygen consumption of the sexual stage-parasites
was relatively low, and it was not different from the asexual blood stage-parasites. However, both stages
of the parasites’ growth and their oxygen consumption were found to be sensitive to atovaquone, cyanide
and 5-fluoroorotate which were inhibitors of mitochondrial electron transport system and pyrimidine bio-
synthetic pathway, respectively. Therefore, the role of mitochondrial organelles with different morpho-
logical properties in the asexual and sexual stages of parasite’s development remains to be elucidated.
KEYWORDS: malaria, Plasmodium falciparum, gametocyte, mitochondria.
ScienceAsia 25 (1999) : 77-83
INTRODUCTION
During the asexual blood stage the human
malarial parasite, Plasmodium falciparum, grows and
matures within the erythrocyte of the human host.
The absence of cristate structure in mitochondrial
organelle at this parasite stage has been demonstrated
from electron microscopic studies.1-5 Energy
requirement is provided by metabolizing glucose
primarily by anaerobic glycolysis.6,7 It has been
clearly evident from biochemical and enzymatic
studies that the asexual stage parasite has
mitochondrial electron transport system and oxygen-
requiring system that is necessary for parasite’s
growth and multiplication.3-10 In addition to the
development of the asexual blood stages, P. falciparum
contains sexual blood stages in the human host
necessary for survival and transmission into the
mosquito vector to complete its life cycle. Relatively
little is known concerning cristate structure and
biochemical function of the mitochondrial organelle
in the sexual blood stage of parasite development,2,11,12
although it develops within the erythrocytes of
human host.
In this communication, the ultrastructural
characteristics of mitochondrial organelles based on
the electron microscopic studies of asexual and
sexual blood stages of P. falciparum grown in vitro
were investigated. The biochemical properties of the
sexual stage parasites were compared to the asexual
blood stage parasites.
MATERIALS AND M ETHODS
Malarial parasites
Human malarial parasite P.falciparum (T9, NP10
and KT3 isolates) was cultivated from the frozen
sample in sorbitol-glycerol cryoprotectant13 by the
candle jar method of Trager and Jensen14, using 5%
hematocrit of human erythrocytes group ‘O’ in the
RPMI 1640 medium supplemented with 25 mM
N-2-hydroxylethylpiperazine-N’-2-ethanesulfonic
acid (Hepes), 32 mM NaHCO3 and 10% human
serum group ‘O’. Cultures started at low parasitemia
(~1-2%) were changed with medium twice daily until
the cultures had ~30% parasitemia and then parasites
were harvested. Synchrony of the culture to get
mainly trophozoite stages was performed by the
sorbitol procedure of Lambros and Vanderberg.15 For
the sexual blood (gametocytic) stages, either NP10
or KT3 isolate was used as gametocyte-producing
strain and then induced according to Ifediba and
Vanderberg.16 Approximately 4% parasitemia of
mixed stages gametocytes were routinely obtained
ESEARCH ARTICLE
R
78 ScienceAsia 25 (1999)
on day 10-15 of cultivation after adding normal fresh
erythrocytes. The gametocytic stages were purified
using Percoll step-wise gradient centrifugation
according to Knight and Sinden.17 Parasites were
freed of their host erythrocytes by incubating in an
equal volume of 0.05% (for sexual gametocytic
stages) or 0.15% saponin (for asexual blood stages)
in the RPMI 1640 medium at 37°C for 20 min, and
washed at least 3 times before experiments.
Micr oscopic examination of P. falcipar um morphology
Transmission electron microscopy (TEM) of the
infected erythrocytes at the asexual and sexual
gametocytic stages was performed according to the
method of Sinden.11 The processed samples were
examined with a JEOL-100SX transmission electron
microscope at the Center of Scientific and Techno-
logical Research Equipment of Chulalongkorn
University, Bangkok. Light microscopy (LM) of the
infected erythrocytes was examined on methanol
fixed and Giemsa’s stained parasites by a Nikon
labophot-2 microscope.
Measur ement of oxygen consumption by P. falcipar um
The rates of oxygen consumption for a consistent
number of host cell-free P. falciparum at both stages
of parasite development were measured in a modified
medium4 containing 75 mM sucrose, 225 nM
mannitol, 5 mM MgCl2, 5 mM KH2PO4, 1 mM
ethylene glycol bis(β-aminoethyl)-N-N-tetraacetic
acid, 5 mM Hepes, pH 7.4, by using a Clark-type
oxygen electrode and YSI oxygen monitor according
to the method of Robinson and Cooper.18 The oxygen
uptake in a chamber with volume of 3 ml was
followed for 3-5 min and recorded at 37°C with a
temperature-controlled circulator. Mitochondrial
inhibitors at desired concentrations were tested
against oxygen consumption of the two stages of the
parasites by injecting into the reacting chamber. The
rates of oxygen consumption by these parasites were
then followed for the next 3-5 min. The 50% effective
concentration (EC50) was defined as the concen-
tration of the inhibitor causing 50% inhibition of the
parasite oxygen consumption, compared to the control
parasite oxygen consumption without inhibitor.
In vitr o antimalarial test
Antimalarial activity against asexual stages of P.
falciparum in vitro (blood schizontocidal activity) was
quantified by measuring % parasitemia in a four-day
culture in the presence of the tested compounds at
various concentrations.19 All compounds were tested
in triplicate at each concentration used. The 50%
inhibitory concentration (IC50) was defined as the
concentration of the compound causing 50%
inhibition of the parasite growth in a 4-day culture,
compared with the compound-free control of the
parasite culture.
Gametocytocidal activity against P. falciparum in
vitro was tested by measuring % parasitemia in a 24-
well microculture plate in the presence of various
concentrations of tested compounds for 2 days
starting at day 5 of cultures after provision of fresh
erythrocytes. At day 7 and again at day 8, all wells
received fresh medium without the compound. Thin
blood films were prepared at day 9. This represents
a four-day period of gametocyte development in
microculture during the antimalarial test, the first
two days under pressure of test compound. The test
method and IC50 value calculation were essentially
from Bhasin and Trager.20
RESULTS AND D ISCUSSION
The in vitro cultures of T9, NP10 and KT3 isolates
(all multidrug-resistant parasites) of P. falciparum
were taken from frozen samples in sorbitol-glycerol
cryoprotectant, and continuously induced for
production of the sexual gametocytic stages for over
2 years. It was noted here that the establishment of
new cultures (or subcultures) by dilution with fresh
erythrocytes markedly reduced gametocyte
production even in the presence of inducing
conditions as described by Ifediba and Vanderberg16,
and this might result in loss of gametocytic stages in
those cultures. T9 parasite was found to be no longer
a gametocyte-producing strain. This may result from
a developmental defect during maturation that has
arisen during the long-term cultivation of the asexual
stages in vitro (~ four-year culture from 1990-1994),
as previously described in the different isolates of P.
falciparum by Guinet et al.21
NP10 and KT3 parasites had been cultured for
more than 2 years and they could be induced for
gametocytogenesis to give parasitemia of the sexual
stages as high as ~4%. The gametocytes were purified
from the asexual stage parasites by the established
method of Percoll step-wise gradient centrifugation.
The purified gametocytes used for TEM and oxygen
consumption are demonstrated in low magnification
of Giemsa’s stained parasites by LM in Fig 1. Mixed
populations of male and female gametocytes of stages
III-V existed in the parasites’ preparations. The
developmental stages of gametocytes were divided
into 5 stages according to the cytological classifica-
tion of Hawking et al.22
ScienceAsia 25 (1999) 79
The mitochondrial organelles observed in P.
falciparum, described as double membranous
structure, were by no means unique between the
asexual and sexual development stages in that they
contain tubular-like cristate structures (Figs 2-5).
The mitochondrial organelles in the asexual blood
stage parasite (Fig 2) were never as numerous as
they were in the sexual gametocyte stage (Figs 4 and
5). Fig. 2 shows a clearly defined double membrane
structure of a mitochondrion in the trophozoite stage
of the asexual blood parasite. Like other protozoan
mitochondria, they are surrounded by a double
membrane, and the inner membrane gives rise to
the tubular cristae. The elongated organelle showed
a limited number of tubular cristae (Fig 3). All
mitochondrial organelles, so far examined, in the
asexual blood stages had no or little internal
membranous whorls rather than well-defined tubular
cristae. They are classified as ‘type I’ mitochondria.
In addition to the unique morphological
characteristics of ‘Type I’ mitochondria in the asexual
stage, the maturing female gametocytic stage parasite
(macrogametocyte) had numerous mitochondrial
organelles (more than 5) containing marked
proliferation and pronounced infolding of the inner
membrane giving rise to large numbers of tubular
cristae with clear intracristal space (Fig 4). They are
classified as ‘Type II’ mitochondria (Figs 6-9). All
macrogametocytes examined had both ‘Type I’ and
‘Type II’ mitochondria (Fig 6). Most mitochondrial
organelles in the macrogametocytes were ‘type II’
mitochondria. Higher magnification of these ‘‘Type
II’ organelles showed them to have different forms
in their shapes of the tubular cristae, for instance,
membranous whorls cristae (Fig 6), or well separated
tubular cristae (Fig 7), or finger-like cristae of closely
folded inner membrane (Figs 8 and 9). By
comparison with the maturing male gametocytic
stage (microgametocyte) (Fig 5), it was noted that
the developing male parasite had less numbers of
the organelles (less than 5), possessed higher
numbers of the cristae, and contained electron-dense
tubular cristae (Figs 10 and 11). They are classified
as ‘Type III’ mitochondria.
Based upon these findings, it is concluded that
1) the male gametocytic parasites have numerous
‘Type III’ mitochondria and their mitochondria are
less numerous than those of the female parasites, 2)
the female parasites contain mainly populations of
‘Type II’ mitochondria and a limited number of
‘Type I’ mitochondria, and 3) the asexual blood
parasite has a single mitochondrion associated with
‘Type I’ organelle (Table 1). Our results reported are
consistent with the observation23 in the other
apicomplexan blood parasite Haemogregarina
myoxocephali at different numbers of mitochondrial
organelles in various developmental stages of its life
cycle: the erythrocytic stage, ~1-2 organelles; the
sexual stage, ~4-6 organelles; and the sporozoites,
~8-10 organelles. The existence of such variations
in mitochondria of male and female gametocytic
stages, both in terms of their numbers and in the
density of cristae, suggests that these developing
sexual stages have high demand for energy
transduction and also metabolic activity differences
from the asexual stages. These active mitochondria
in the sexual stages may be necessary for their
survival during transmission into the mosquito
vector.
To see whether the mitochondria in sexual
gametocytic stages of P. falciparum were biochemically
active or not, mitochondrial oxygen consumption
of the parasites were performed with known
mitochondrial inhibitors. It was found that
mitochondrial oxygen consumption of cultured P.
falciparum from both stages were not different (Table
2). They had relatively low activity (~ 200 - 240
nmol/min/108 parasites), compared to the human
leukocytes which had an oxygen consumption of
1,090 nmol/min/108 cells (n=2). Cyanide, a known
inhibitor of mammalian electron transporting
complex IV (cytochrome c oxidase),24 had an
inhibitory effect against oxygen consumption of the
host cell-free parasites isolated from both asexual
and sexual stages (Table 2). At a concentration of
1.0x 10-3 M, cyanide inhibited 52% and 88% of the
oxygen consumption by the asexual and sexual stage
parasites, respectively. Cyanide has been shown to
exhibit low antimalarial effect against P. falciparum
growth in vitro with IC50 value in the micromolar
level.8,25 It has inhibitory effects against the purified
cytochrome c oxidase of both P. berghei4 and P.
falciparum.10
The parasite oxygen consumption of both stages
was found to be sensitive to atovaquone inhibition
Table 1. The comparison of type and number of
mitochondrial organelle between asexual and
sexual blood stages of
P. falciparum
.
Stages of parasite Mitochondrial characteristics
Type Number
Asexual stage I 1
Sexual stage
Female II >>I >5
Male III <5
80 ScienceAsia 25 (1999)
Fig 1 Light micrograph of purified gametocytes from in vitro
cultures of P. falciparum. All preparations were methanol
fixed and stained with Giemsa’s stain. The bar represents
10 µM.
Fig 2 Transmission electron micrograph (TEM) of the asexual
trophozoite stage of P.falciparum. The mitochondrion shows
a clearly defined double membranes and an elogated form
which is prepared for binary fission. A membrane whorls-
like tubular crista resulting from an underdeveloped inner
membrane is marked by an arrowhead. The bar represents
1 µM. N, nucleus; M, mitochondria; P, crystalline pigment.
Fig 3 TEM of higher magnification of ‘Type I’ mitochondria in
the asexual trophozoite stage of P. falciparum. Only one
tubular cristate structure is demonstrated (arrowhead). The
bar represents 1 µM. M, mitochondria; P, crystalline
pigment.
Fig 5 TEM of maturing male gametocytic stage (microgame-
tocyte in stage IV) of P. falciparum. Few ‘Type III’
mitochondria are observed, Laveran’s ‘Bib’ is also visible
at the middle left. The bar represents 1µM. N, nucleus; M,
mitochondria; P, crystalline pigment.
Fig 4 TEM of maturing female gametocytic stage (macrogame-
tocyte in stage IV) of P. falciparum. Numerous ‘Type II’
mitochondria are observed. The bar represents 1 µM. M,
mitochondria; P, crystalline pigment.
Fig 6 A high magnification TEM of mitochondria in a maturing
female gametocyte. The lower (Type I) and the upper (Type
II) mitochondria are shown. The tubular cristae of ‘Type
II’ organelle is marked by an arrowhead. The bar represents
1 µM. M, mitochondria; P, crystalline pigment.
ScienceAsia 25 (1999) 81
Fig 7 TEM of ‘Type II’ mitochondria in a female gametocyte. Two
organelles are observed with high magnification. They
contain numerous tubular cristae, marked by arrowhead.
The bar represents 1 µM. M, mitochondria.
Fig 8 TEM of variant for ms of ‘Type II’ mitochondria in a female
gametocyte. Three organelles are observed with high
magification. Numerous tubular cristae resulting from the
extensively folding of the inner membrane are observed.
An arrowhead indicates the finger-like cristae. The bar
represents 1 µM. M, mitochondria; P, crystalline pigment.
Fig 9 TEM of another variant for ms of ‘Type II’ mitochondria in
a female gametocyte. The finger-like cristae is typically
found in the female gametocytes (arrowhead). The bar
represents 1 µM. M, mitochondria; P, crystalline pigment.
Fig 11 TEM of ‘‘Type III’mitochondria in a male gametocyte. An
apicoplast is observed with a multi-membranous organelle
containing electron-dense matrix and absence of internal
cristae. The bar represents 1 µM. M, mitochondria; P,
crystalline pigment. A, apicoplast.
Fig 10 TEM of ‘Type III’mitochondria in a male gametocyte.
Electron-dense and finely compact of the tubular cristae
is typically associated with this type of the organelle. The
numbers of cristae in ‘type III’ mitochondria are more than
those of ‘‘Type II’ mitochondria of the female gametocytes.
The bar represents 1 µM. An arrowhead indicates electron-
dense tubular cristae. M, mitochondria.
82 ScienceAsia 25 (1999)
(Tables 2 and 3). The antimalarial drug atovaquone
is a mitochondrial inhibitor of the parasite electron
transporting complex III (ubiquinol-cytochrome c
reductase).10,26-28 Furthermore, the atovaquone had
moderate gametocytocidal activity with IC50 of 5x10-8
M, compared to its potent blood schizontocidal
activity (IC50=5x10-10 M). It has been recently shown
that atovaquone is indeed a gametocytocidal drug.29
These lines of evidence would provide mitochondria
in the sexual gametocytic stages as a possible
chemotherapeutic target.
Our results suggest that the abundant mitochon-
dria in the sexual gametocytic stage parasites were
still in the underdeveloped forms but biochemically
active at least with regards to oxygen consumption.
The mitochondrial heterogeneity also suggests their
role in energy production. Mitochondrial ATP
synthetase inhibitors are reported to have
antimalarial activity against the asexual growth of P.
falciparum at micromolar concentrations.8,25,30
Existence of ATP synthetase activity which is respon-
sible for ATP production in the sexual gametocytic
stage remains to be elucidated.
The findings on the relatively low oxygen
consumption and reduced sensitivity to cyanide
(Tables 2 and 3) suggest that P. falciparum operates
either a branched chain respiratory pathway
containing a non-cytochrome electron transport
system with an alternative oxidase which is
insensitive to cyanide6,31 or a branched electron
transport chain consisting of a specialized
cytochrome system in which fumarate acts as an
electron acceptor and is reduced to succinate by an
NADH-dependent fumarate reductase.3 The latter
pathway has been described in the adult round worm
Ascaris suum showing that ATP is anaerobically
produced by substrate level phosphorylation in the
branched electron transport pathway.32
More interestingly, 5-fluoroorotate is reported to
be a potent inhibitor of pyrimidine biosynthetic
pathway of P.falciparum.19,33 It showed marked
inhibitory effect on the oxygen consumption by both
asexual and sexual stage parasites (Table 3),
suggesting that the parasite in the sexual stage has
linkage of the two metabolic pathways, pyrimidine
biosynthesis and mitochondrial electron transport
system, through dihydroorotate dehydrogenase. The
association between the pyrimidine pathway and
mitochondrial electron transport system in the
asexual stage parasite has been confirmed.5,9 It is then
concluded that P. falciparum in both developmental
stages have functional mitochondria that contribute
significantly to de novo synthesis of pyrimidine and
to the energy metabolism of the parasite.
Whether or not the mitochondrial heterogeneity
observed in the parasites have functional significance
must await study of their biochemistry and
physiology, for instance, enzymatic activities of the
mitochondrial electron transport chain in the sexual
stage parasite, differences in the mechanism of
energy metabolism, mitochondrial ATP synthetase,
and role of oxygen tension on gametocytes
circulating in human blood system.
ACKNOWLEDGEMENTS
We thank S Vettchagarun and D Burat for their
dedicated technical assistance with EM techniques
and some experiments on oxygen uptake. The
parasites T9 and KT3 isolates were kindly provided
by S Thaitong of Chulalongkorn University and
P Petmitr of Mahidol University, respectively. This
work was supported by the UNDP/World Bank/
WHO Special Programme for Research and Training
in Tropical Diseases. J Krungkrai is a career
development award recipient from the National
Science and Technology Development Agency of
Thailand.
Table 2. The mitochondrial oxygen consumption by
P.
falciparum
at asexual and sexual stages of
development.
Stages of parasite
Oxygen consumption a
(nmol/min/10 8 parasites)
Control +Atovaquone b+Cyanide c
Asexual stage 204±16 10±2 97±10
Sexual stage 242±20 126±14 30±4
aValues are means ±SD, taken from 3-4 separate experiments of the parasite
preparations.
bFinal concentration used was 1x10-6M atovaquone.
cFinal concentration used was 1x10-3M potassium cyanide.
Table 3. The 50% effective concentrations (EC50) of
mitochondrial electron transport system and
pyrimidine biosynthetic pathway inhibitors on
parasite oxygen consumption.
Stages of parasite EC50 (M)
Atovaquone Cyanide 5-Fluor oorotate
Asexual stage 5x10-8 9x10-4 1x10-7
Sexual stage 9x10-7 2x10-4 5x10-7
ScienceAsia 25 (1999) 83
REFERENCES
1. Aikawa M, Huff CG, and Sprinz H (1966) Comparative feeding
mechanisms of avian and primate malarial parasites. Mil Med
131, 969-83.
2. Langreth SG, Jensen JB, Reese RT and Trager W (1978) Fine
structure of human malaria in vitro. J Protozool 25, 443-52.
3. Fry M and Beesley JE (1991) Mitochondria of mammalian
Plasmodium spp. Parasitology 102, 17-26.
4. Krungkrai J, Krungkrai SR and Bhumiratana A (1993)
Plasmodium berghei: partial purification and characterization
of the mitochondrial cytochrome c oxidase. Exp Parasitol 77,
136-46.
5. Krungkrai J (1995) Purification, characterization and
localization of mitochondrial dihydroorotate dehydrogenase
in Plasmodium falciparum, human malaria parasite. Biochim
Biophys Acta 1243, 351-60.
6. Sherman IW (1979) Biochemistry of Plasmodium (malaria
parasites). Microbiol Rev 43 453-95.
7. Scheibel LW (1988) Plasmodial metabolism: carbohydrate. In
“Malaria” ( WH Wernsdorfer and I McGregor, Eds), Vol I, pp
171-217. Churchill Livingstone, New York.
8. Ginsburg H, Divo AA, Geary TG, Boland MT, and Jensen JB
(1986) Effect of mitochondrial inhibitors on intraerythrocytic
intraerythrocytic Plasmodium falciparum in in vitro cultures. J
Protozool 33, 121-5.
9. Krungkrai J, Cerami A and Henderson GB (1991) Purification
and characterization of dihydroorotate dehydrogenase from
the rodent malaria parasite Plasmodium berghei. Biochemistry
30, 1934-9.
10. Krungkrai J, Krungkrai SR, Suraveratum N, and Prapunwattana
P (1997) Mitochondrial ubiquinol-cytochrome c reductase and
cytochrome c oxidase: chemotherapeutic targets in malarial
parasites. Biochem Mol Biol Inter 42, 1007-14.
11.Sinden RE (1982) Gametocytogenesis of Plasmodium
falciparum in vitro: an electron microscopic study. Parasitology
84, 1-11.
12.Petmitr S and Krungkrai J (1995) Mitochondrial cytochrome
b gene in two developmental stages of human malarial parasite
Plasmodium falciparum. Southeast Asian J Trop Med Public
Health 26, 600-5.
13.Trigg PI (1978) Plasmodiidae. In ‘‘Methods of Culturing
Parasites in vitro” (AER Taylor and JR Baker Eds), pp 89-100.
Academic Press, London.
14.Trager W and Jensen JB (1976) Human malaria parasites in
continuous culture. Science 193, 673-5.
15.Lambros C and Vanderberg JP (1979) Synchronization of
Plasmodium falciparum erythrocytic stages in culture. J
Parasitol 65, 418-20.
16. Ifediba T and Vanderberg JP (1981) Complete in vitro maturation
of Plasmodium falciparum gametocytes. Nature 294, 364-6.
17.Knight A and Sinden RE (1982) The purification of
gametocytes of Plasmodium falciparum and P yoelii nigeriensis
by colloidal silica (Percoll) gradient centrifugation. Trans R
Soc Trop Med Hyg. 76, 503-9.
18.Robinson J and Cooper JM (1970) Method of determining
oxygen concentrations in biological media, suitable for
calibration of the oxygen electrode. Anal Biochem 33, 390-9.
19.Krungkrai J, Krungkrai SR and Phakanont K (1992) Anti-
malarial activity of orotate analogs that inhibit dihydroorotase
and di-hydroorotate dehydrogenase. Biochem Pharmacol 43,
1295-301.
20.Bhasin VK and Trager W (1987) Gametocytocidal effects in
vitro of primaquine and related compounds on Plasmodium
falciparum. In “Primaquine: pharmacokinetics, metabolism,
toxicity, and activity” (WH Wernsdorfer and PI Trigg, Eds),
pp 145-53. John Wiley and Sons, Chichester, New York.
21.Guinet F, Dvorak JA, Fujioka H, Keister DB, Muratova O,
Kaslow DC, Aikawa M, Vaidya AB and Wellems TE (1996) A
developmental defect in Plasmodium falciparum male
gametogenesis. J Cell Biol 135, 269-78.
22.Hawking F, Wilson ME and Gammage K (1971) Evidence for
cyclic development and short-lived maturity in the
gametocytes of Plasmodium falciparum. Trans R Soc Trop Med
Hyg 65, 549-59.
23.Siddall ME and Desser SS (1992) Ultrastructure of
gametogenesis and sporogony of Haemogregarina (sensu lato)
myoxocephali (Apicomplexa: Adeleina) in the marine leech
Malmiana scorpii. J Protozool 39, 545-54.
24. Hatefi Y (1985) The mitochondrial electron transport and oxida-
tive phosphorylation system. Annu Rev Biochem 54, 1015-69.
25. Krungkrai J, Learngaramkul P, Kanjanarithisak R and Krungkrai
SR (1994) Mitochondrial function of human malarial parasite
Plasmodium falciparum cultivated in vitro: implications for
antimalarial drug design. Chula Med J 38, 315-23.
26. Fry M and Pudney M (1992) Site of action of the antimalarial
hydroxynaphoquinone, 2-[trans-4-(4’-
chlorphenyl)cyclohexyl]-3-hydroxy-1,4-naphthoquinone
(566C80). Biochem Pharamacol 43, 1545-53.
27. Vaidya AB, Lashgari MS, Pologe LG and Morrisey J (1993)
Structural features of Plasmodium cytochrome b that may
underline susceptibility to 8-aminoquinolines and hydroxy-
naphthoquinones. Mol Biochem Parasitol 58, 33-42.
28. Srivastava IK, Rottenberg H and Vaidya AB (1997) Atovaquone,
a broad spectrum antiparasitic drug, collapses mitochondrial
membrane potential in a malarial parasite. J Biol Chem 272,
3961-6.
29.Fleck SL, Pudney M and Sinden RE (1996) The effect of
atovaquone (566C80) on the maturation and viability of
Plasmodium falciparum gametocytes in vitro. Trans R Soc Trop
Med Hyg 90, 309-12.
30. Basco LK and LeBras J (1994). In vitro activity of mitochondrial
ATP synthetase inhibitors against Plasmodium falciparum. J
Euk Microbiol 41, 179-83.
31.Murphy AD, Daeller JE, Hearn B and Lang-Unnasch N (1997)
Plasmodium falciparum: cyanide-resistant oxygen consumption.
Exp Parasitol 87, 112-20.
32.Kita K (1992) Electron-transfer complexes of mitochondria
in Ascaris suum. Parasitol Today 8, 155-9.
33.Seymour KK, Lyons SD, Phillips L, Rieckmann KH and
Christopherson RI (1994) Cytotoxic effects of inhibitors of de
novo pyrimidine biosynthesis upon Plasmodium falciparum.
Biochemistry 33, 5268-74.
... Antibiogram profiles of the bacterial isolates suggest that all of them belong to MDR strains. The presence of these bacteria in hospital wards indicated that they could constitute a major health risk to humans and associated animals like cats and rodents because urinary tract, biliary tract and peritoneal infections, wound infections, sepsis, gastroenteritis, and pneumonia are all possible illnesses caused by these microbes [32,33]. ...
Article
Full-text available
German cockroaches, Blattella germanica L. are the most dangerous pests in hospital environments which cause diseases like food poisoning, dysentery, and diarrhea. Antibiogram and multidrug resistance (MDR) patterns of the bacterial isolates from the cockroaches inhabiting three Wards of Rajshahi Medical College Hospital, Rajshahi, Bangladesh, were determined. A total of 52 bacterial isolates, 26 from the cuticle and 26 from the gut, comprising 20 (38.5 %) from the Neuromedicine ward, 18 (34.6 %) from the Orthopaedic ward, and 14 (26.9 %) from the Surgery ward, were used for biochemical assays and diagnostic characteristics, where seven Gram-negative rod species were identified. E. coli (n=8 or 40 %), Klebsiella spp. (n=5 or 28 %) and Klebsiella spp. (5 or 35 %) constituted the most dominant bacteria from the three wards respectively. Antibiogram of the bacterial isolates against 18 commonly used antibiotics revealed that Serratia, Klebsiella, Escherichia, and Pseudomonas were resistant to Ceftazidime, whereas Salmonella, Enterobacter, and Proteus were sensitive to the drug. Results of the present study suggest that the German cockroaches might serve as reservoirs for pathogenic and MDR bacteria, which in turn could be responsible for the spread of common food-borne diseases in hospital patients, their attendants, and health professionals.
Chapter
Nine of the 20 most utilized freshwater aquaculture fish species around the world have been genetically modified (GM), and another five species of aquaculture interest and two species for research are being extensively worked on. Modifications are focused on production-related traits, with growth enhancement by far in the most advanced state with respect to commercial application currently awaiting regulatory decision. Other aspects more or less developed are phenotypic modifications of ornamental fishes and, for disease resistance, biomonitoring, biocontrol and pharmaceutical production and xenotransplantation of tissue to humans. While GM technology may hold potential benefits for the fresh water ecosystem, they are not viewed scientifically as universally positive, and a number of critical issues have been identified, including ecological risks, food safety and ethics. More specifically, GM fishes may pose problems in making use of limited fresh water, pollution and disrupting the ecological balance. Such issues will need to be resolved before commercial application will become widely accepted. Assessing ecological risk is complicated by pleiotropic effects from the transgene acting on the animals, the sensitivity to rearing conditions and the complexity of natural freshwater ecosystems.
Article
Sporozoites of the apicomplexan Cryptosporidium parvum possess a small, membranous organelle sandwiched between the nucleus and crystalloid body. Based upon immunolabelling data, this organelle was identified as a relict mitochondrion. Transmission electron microscopy and tomographic reconstruction reveal the complex arrangement of membranes in the vicinity of this organelle, as well as its internal organization. The mitochondrion is enveloped by multiple segments of rough endoplasmic reticulum that extend from the outer nuclear envelope. In tomographic reconstructions of the mitochondrion, there is either a single, highly-folded inner membrane or multiple internal subcompartments (which might merge outside the reconstructed volume). The infoldings of the inner membrane lack the tubular "crista junctions" found in typical metazoan, fungal, and protist mitochondria. The absence of this highly conserved structural feature is congruent with the loss, through reductive evolution, of the normal oxidative phosphorylation machinery in C. parvum. It is proposed that the retention of a relict mitochondrion in C. parvum is a strategy for compartmentalizing away from the cytosol toxic ferrous iron and sulfide, which are needed for iron sulfur cluster biosynthesis, an essential function of mitochondria in all eukaryotes.
Article
The mitochondrial electron transport system is necessary for growth and survival of malarial parasites in mammalian host cells. NADH dehydrogenase of respiratory complex I was demonstrated in isolated mitochondrial organelles of the human parasite Plasmodium falciparum and the mouse parasite Plasmodium berghei by using the specific inhibitor rotenone on oxygen consumption and enzyme activity. It was partially purified by two sequential steps of fast protein liquid chromatographic techniques from n-octyl glucoside solubilization of the isolated mitochondria of both parasites. In addition, physical and kinetic properties of the malarial enzymes were compared to the host mouse liver mitochondrial respiratory complex I either as intact or as partially purified forms. The malarial enzyme required both NADH and ubiquinone for maximal catalysis. Furthermore, rotenone and plumbagin (ubiquinone analog) showed strong inhibitory effect against the purified malarial enzymes and had antimalarial activity against in vitro growth of P. falciparum. Some unique properties suggest that the enzyme could be exploited as chemotherapeutic target for drug development, and it may have physiological significance in the mitochondrial metabolism of the parasite.
Article
Plasmodium falciparum can now be maintained in continuous culture in human erythrocytes incubated at 38°C in RPMI 1640 medium with human serum under an atmosphere with 7 percent carbon dioxide and low oxygen (1 or 5 percent). The original parasite material, derived from an infected Aotus trivirgatus monkey, was diluted more than 100 million times by the addition of human erythrocytes at 3- or 4-day intervals. The parasites continued to reproduce in their normal asexual cycle of approximately 48 hours but were no longer highly synchronous. They have remained infective to Aotus.
Article
ABSTRACT The sexual and sporogonic development of Haemogregarina (sensu lato) myoxocephali, an apicomplexan blood parasite of longhorn sculpin, Myoxocephalus octodecemspinosus, was studied by transmission electron microscopy. All stages of development were observed epicellularly within intestinal epithelial cells of the leech Malmiana scorpii. During microgametogenesis nuclear division was characterized by a transnuclear cytoplasmic channel containing the spindle microtubules. Four aflagellate microgametes were formed. During fertilization, a single microgamete nucleus was associated with the endoplasmic reticulum in the macrogamete, followed by fusion of the nuclear envelopes of the gametes. Sporogony involved peripheral budding of sporozoite anlagen and subsequent development to form approximately 32 sporozoites in mature oocysts.
Article
Intraerythrocytically, bird and mammalian malarias appear to derive energy by metabolizing glucose to lactic acid via a conventional pathway of anaerobic glycolysis. If supplied with oxygen, avian parasites may oxidize a portion of the pyruvate to CO2 and water by means of the citric acid cycle, whereas rodent and primate malarias, which lack a functional citric acid cycle, are unable to do so and have lactate as their primary end product. Erythrocyte-free P. gallinaceum cells produce appreciable quantities of acetate from glucose and pyruvate. Since plasmodia are unable to synthesize CoA de novo, it is possible that in P. gallinaceum acetate formation is due to a lack of host-supplied CoA. In P.knowlesi it may be that volatile acids are formed by a pyruvate clastic reaction, but the enzymes involved (if they exist) have not been looked for. However, in view of the fragile nature of free parasites, acetate and formate production may reflect deranged metabolism due to in situ leakiness of plasmodia or may be a consequence of insult during the isolation procedure. There is no evidence for a pentose phosphate shunt in malarial parasites since the first enzyme in the pathway (G6PDH) is absent. Indeed, the only enzyme in this pathway that has been identified consistently is 6-phosphogluconate dehydrogenase. Lacking a pentose shunt, the parasites must have other means for obtaining ribose and reducing NADP. It has been suggested that action by phosphorylases supplies the pentoses and that glutamic dehydrogenase provides for the reduction of NADP. Evidence for an energy-yielding electron transport chain is at best circumstantial. In all of the malarias studied, the only enzyme found to be associated with this system is cytochrome oxidase. It is conceivable that in the acristate rodent malarias and in P. knowlesi, and perhaps even in those malarias having cristate mitochondria (avians and P. falciparum), cytochrome oxidase is involved in the de novo pyrimidine biosynthetic pathway and not in energy-yielding reactions. Malarial parasites are incapable of de novo purine biosynthesis; however, pyrimidines are synthesized de novo. Exogenously supplied purines and orotic acid are transported and incorporated by infected erythrocytes and plasmodia, whereas pyrimidines (uracil and thymidine) are not. There is evidence to support the contention that hypoxanthine is the preferred purine of the parasites in vivo and that it is derived from the catabolism of erythrocytic ATP. Plasmodia have a distinctive DNA and rRNA base composition. Malarial parasite ribosomes are not provided for by host cell ribosomal subparticles, and the mechanism of protein synthesis by the parasites is typically eucaryotic. The capacity of the parasites for de novo amino acid biosynthesis is limited, and it appears that host cell hemoglobin provides most of the amino acids. For some species, isoleucine and methionine must be supplied exogenously for good plasmodial growth. The degradation of erythrocyte hemoglobin by parasite proteases leaves a golden brown-black residue called hemozoin (malarial pigment). Hemozoin consists of insoluble monomers and dimers of hematin, methemoglobin, and ferriprotoporphyrin coupled to plasmodial protein. The functional significance of hemozoin is not completely understood. The only well-characterized plasmodial protein is the HRP of P. lophurae. It is possible that HRP is localized in the polar organelles of the merozoites and is involved in the process of invasion. Information regarding the vitamin requirements of malarial parasites is scanty. Plasmodia are incapable of synthesizing CoA from pantothenate and rely on the host cell for this cofactor; this may be one reason for their being obligate intracellular parasites. By contrast, Plasmodium can synthesize folate from pABA.
Article
The erythrocytic cycle of the human malaria parasite, Plasmodium, falciparum, was examined by electron microscopy. Three strains of parasites maintained in continuous culture in human erythrocytes were compared with in vivo infections in Aotus monkeys. The ultrastructure of P. falciparum is not altered by continuous cultivation in vitro. Mitochondria contain DNA-like filaments and some cristae at all stages of the erythrocytic life cycle. The Golgi apparatus is prominent at the schizont stage and may be involved in the formation of rhoptries. In culture, knob-like protrusions first appear on the surface of trophozoite-infected erythrocytes. The time of appearance of knobs on cells in vitro correlates with the life cycle stage of parasites which are sequestered from the peripheral circulation in vivo. Knob material of older parasites coalesces and forms extensions from the erythrocyte surface. Some of this material is sloughed from the host cell surface. The parasitophorous vacuole membrane breaks down in erythrocytes containing mature merozoites both in vitro and in vivo. Merozoite structure is similar to that of P. knowlesi. The immature gametocytes in culture have no knobs.
Article
Synchronous development of the erythrocytic stages of a human malaria parasite, Plasmodium falciparum, in culture was accomplished by suspending cultured parasites in 5% D-sorbitol and subsequent reintroduction into culture. Immediately after sorbitol treatment, cultures consisted mainly of single and multiple ring-form infections. At the same time, varying degrees of lysis of erythrocytes infected with the more mature stages of the parasite was evident. Approximately 95% of the parasites were in the ring stage of development at 48 and 96 hr after sorbitol treatment-likewise, a high percentage of trophozoite and schizont stages was observed at 24, 72, and 120 hr. D-Mannitol produced similar, selective, lytic effects.
Article
Plasmodium falciparum can now be maintained in continuous culture in human erythrocytes incubated at 38 degrees C in RPMI 1640 medium with human serum under an atmosphere with 7 percent carbon dioxide and low oxygen (1 or 5 percent). The original parasite material, derived from an infected Aotus trivirgatus monkey, was diluted more than 100 million times by the addition of human erythrocytes at 3- or 4-day intervals. The parasites continued to reproduce in their normal asexual cycle of approximately 48 hours but were no longer highly synchronous. The have remained infective to Aotus.
Article
The site of action of the antimalarial compound 2-[trans-4-(4'-chlorophenyl) cyclohexyl]-3-hydroxy-1,4-naphthoquinone (566C80), would appear to be the mitochondrial respiratory chain. Studies reported herein have demonstrated 566C80 to be a potent and selective mitochondrial inhibitor with mitochondria isolated from Plasmodium falciparum and P. yoelii. Selective assay of individual respiratory chain complexes has shown the primary site of action of 566C80 to be the cytochrome bc1 complex (Complex III): supportive evidence from difference spectroscopy indicates the site of inhibition to lie between cytochromes b and c1 of this complex. Using [14C]566C80, evidence is presented which suggests that 566C80 may become irreversibly bound to a polypeptide with an approximate molecular mass of 11,500 Da.
Article
Dihydroorotase and dihydroorotate dehydrogenase, two enzymes of the pyrimidine biosynthetic pathway, were purified from Plasmodium berghei to apparent homogeneity. Orotate and a series of 5-substituted derivatives were found to inhibit competitively the purified enzymes from the malaria parasite. The order of effectiveness as inhibitors on pyrimidine ring cleavage reaction for dihydroorotase was 5-fluoro orotate greater than 5-amino orotate, 5-methyl orotate greater than orotate greater than 5-bromo orotate greater than 5-iodo orotate with Ki values of 65, 142, 166, 860, 2200 and greater than 3500 microM, respectively. 5-Fluoro orotate and orotate were the most effective inhibitors for dihydroorotate dehydrogenase. In vitro, 5-fluoro orotate and 5-amino orotate caused 50% inhibition of the growth of P. falciparum at concentrations of 10 nM and 1 microM, respectively. In mice infected with P. berghei, these two orotate analogs at a dose of 25 mg/kg body weight eliminated parasitemia after a 4-day treatment, an effect comparable to that of the same dose of chloroquine. The infected mice treated with 5-fluoro orotate at a lower dose of 2.5 mg/kg had a 95% reduction in parasitemia. The effects of the more potent compounds tested in combination with inhibitors of other enzymes of this pathway on P. falciparum in vitro and P. berghei in vivo are currently under investigation. These results suggest that the pyrimidine biosynthetic pathway in the malarial parasite may be a target for the design of antimalarial drugs.
Article
Dihydroorotate dehydrogenase (DHODase) has been purified 400-fold from the rodent malaria parasite Plasmodium berghei to apparent homogeneity by Triton X-100 solubilization followed by anion-exchange, Cibacron Blue F3GA-agarose affinity, and gel filtration chromatography. The purified enzyme has a molecular mass of 52 +/- 2 kDa on sodium dodecyl sulfate-polyacrylamide gel electrophoresis and of 55 +/- 6 kDa by gel filtration chromatography, and it has a pI of 8.2. It is active in monomeric form, contains 2.022 mol of iron and 1.602 acid-labile sulfurs per mole of enzyme, and does not contain a flavin cofactor. The purified DHODase exhibits optimal activity at pH 8.0 in the presence of the ubiquinone coenzyme CoQ6, CoQ7, CoQ9, or CoQ10. The Km values for L-DHO and CoQ6 are 7.9 +/- 2.5 microM and 21.6 +/- 5.5 microM, respectively. The kcat values for both substrates are 11.44 min-1 and 11.70 min-1, respectively. The reaction product orotate and an orotate analogue, 5-fluoroorotate, are competitive inhibitors of the enzyme-catalyzed reaction with Ki values of 30.5 microM and 34.9 microM, respectively. The requirement of the long-chain ubiquinones for activity supports the hypothesis of the linkage of pyrimidine biosynthesis to the electron transport system and oxygen utilization in malaria by DHODase via ubiquinones [Gutteridge, W. E., Dave, D., & Richards, W. H. G. (1979) Biochim. Biophys. Acta 582, 390-401].