ArticlePDF Available

Nitric Oxide-Mediated Proteasome-Dependent Oligonucleosomal DNA Fragmentation in Leishmania amazonensis Amastigotes

American Society for Microbiology
Infection and Immunity
Authors:

Abstract and Figures

Resistance to leishmanial infections depends on intracellular parasite killing by activated host macrophages through the l-arginine-nitric oxide (NO) metabolic pathway. Here we investigate the cell death process induced by NO for the intracellular protozoan Leishmania amazonensis. Exposure of amastigotes to moderate concentrations of NO-donating compounds (acidified sodium nitrite NaNO2 or nitrosylated albumin) or to endogenous NO produced by lipopolysaccharide or gamma interferon treatment of infected macrophages resulted in a dramatic time-dependent cell death. The combined use of several standard DNA status analysis techniques (including electrophoresis ladder banding patterns, YOPRO-1 staining in flow cytofluorometry, and in situ recognition of DNA strand breaks by TUNEL [terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling] assay) revealed a rapid and extensive fragmentation of nuclear DNA in both axenic and intracellular NO-treated amastigotes of L. amazonensis. Despite some similarities to apoptosis, the nuclease activation responsible for characteristic DNA degradation was not under the control of caspase activity as indicated by the lack of involvement of cell-permeable inhibitors of caspases and cysteine proteases. In contrast, exposure of NO-treated amastigotes with specific proteasome inhibitors, such as lactacystin or calpain inhibitor I, markedly reduced the induction of the NO-mediated apoptosis-like process. These data strongly suggest that NO-induced oligonucleosomal DNA fragmentation in Leishmania amastigotes is, at least in part, regulated by noncaspase proteases of the proteasome. The determination of biochemical pathways leading up to cell death might ultimately allow the identification of new therapeutic targets.
This content is subject to copyright. Terms and conditions apply.
INFECTION AND IMMUNITY, July 2002, p. 3727–3735 Vol. 70, No. 7
0019-9567/02/$04.000 DOI: 10.1128/IAI.70.7.3727–3735.2002
Copyright © 2002, American Society for Microbiology. All Rights Reserved.
Nitric Oxide-Mediated Proteasome-Dependent Oligonucleosomal
DNA Fragmentation in Leishmania amazonensis Amastigotes
Philippe Holzmuller,
1
Denis Sereno,
1
Mireille Cavaleyra,
1
Isabelle Mangot,
1
Sylvie Daulouede,
2
Philippe Vincendeau,
2
and Jean-Loup Lemesre
1
*
UR 008 Pathogénie des Trypanosomatidés, Institut de Recherche pour le Développement, 34032 Montpellier Cedex 1,
1
and
Laboratoire de Parasitologie, Université Victor Segalen Bordeaux II, 33076 Bordeaux Cedex,
2
France
Received 4 September 2001/Returned for modification 15 November 2001/Accepted 21 March 2002
Resistance to leishmanial infections depends on intracellular parasite killing by activated host macrophages
through the L-arginine–nitric oxide (NO) metabolic pathway. Here we investigate the cell death process
induced by NO for the intracellular protozoan Leishmania amazonensis. Exposure of amastigotes to moderate
concentrations of NO-donating compounds (acidified sodium nitrite NaNO
2
or nitrosylated albumin) or to
endogenous NO produced by lipopolysaccharide or gamma interferon treatment of infected macrophages
resulted in a dramatic time-dependent cell death. The combined use of several standard DNA status analysis
techniques (including electrophoresis ladder banding patterns, YOPRO-1 staining in flow cytofluorometry, and
in situ recognition of DNA strand breaks by TUNEL [terminal deoxynucleotidyltransferase-mediated dUTP-
biotin nick end labeling] assay) revealed a rapid and extensive fragmentation of nuclear DNA in both axenic
and intracellular NO-treated amastigotes of L. amazonensis. Despite some similarities to apoptosis, the
nuclease activation responsible for characteristic DNA degradation was not under the control of caspase
activity as indicated by the lack of involvement of cell-permeable inhibitors of caspases and cysteine proteases.
In contrast, exposure of NO-treated amastigotes with specific proteasome inhibitors, such as lactacystin or
calpain inhibitor I, markedly reduced the induction of the NO-mediated apoptosis-like process. These data
strongly suggest that NO-induced oligonucleosomal DNA fragmentation in Leishmania amastigotes is, at least
in part, regulated by noncaspase proteases of the proteasome. The determination of biochemical pathways
leading up to cell death might ultimately allow the identification of new therapeutic targets.
Leishmania spp. are obligate intracellular protozoan para-
sites of macrophages that are responsible for a wide range of
human diseases, including self-healing skin lesions, diffuse cu-
taneous and mucosal lesions, or fatal visceral infections. In the
Leishmania life cycle, two principal parasite forms exist: (i) the
amastigote form that develops inside mononuclear phagocytes
of a vertebrate host and (ii) the motile promastigote form that
develops in the vector gut. The various possible outcomes of
leishmanial infection have been associated with expansion of
specific T helper lymphocyte populations (52). Immune control
of leishmaniasis involves a dominant Th1 response, leading to
macrophage activation and elimination of intracellular para-
sites through the induction of nitric oxide synthase (NOS II)
and NO synthesis from L-arginine. This prototypical model has
been largely evidenced in murine leishmaniasis (17, 22, 33).
Human activated macrophages can also induce antileishmanial
activity via the L-arginine NO pathway (44, 58, 60). Studies on
human cutaneous leishmaniasis reveal that Leishmania killing
is associated with NO production (40). Moreover, the capacity
of canine macrophages to eliminate intracellular amastigotes
through a NO-dependent mechanism has been documented
(46, 55, 59).
NO-dependent cytostatic and/or cytotoxic activities by acti-
vated macrophages on various parasites have been clearly
demonstrated (20, 26, 29, 33, 57). In Leishmania infections,
NO-generating creams applied topically to lesions revealed a
modest efficacy in mouse models, whereas they were able to
cure human patients (15, 65). Moreover, studies have shown
that successful chemotherapy in a murine and canine model of
visceral leishmaniasis depended on upregulation of the L-argi-
nine NO pathway (14, 58). However, the cellular and molecu-
lar mechanisms whereby NO exerts its cytotoxic activity are not
yet well understood. NO toxicity results from complex phe-
nomena involving several molecular targets. In antitumoral
models, impairment of essential cellular functions, including
mitochondrial respiration, DNA synthesis by blocking ribonu-
cleotide reductase (31), and enzyme inactivation through Fe/S
center alterations or S nitrosylation, have been documented
(16, 19). Recent studies found that several parasite targets may
be affected by NO toxicity in Leishmania parasites. These tar-
gets include metabolic enzymes, such as GAPDH (glyceralde-
hyde-3-phosphate dehydrogenase) and aconitase (29, 32) or
cysteine proteinase (51).
Apoptosis represents an important mechanism by which ex-
ogenous or endogenous NO mediates toxicity in mammalian
cells (1, 36, 41, 42, 63). We investigated the genomic DNA
status of NO-treated Leishmania amazonensis amastigotes.
Several apoptosis detection methods (appearance of the char-
acteristic DNA ladder banding pattern on agarose gels and in
situ TUNEL [terminal deoxynucleotidyltransferase-mediated
dUTP-biotin nick end labeling] and flow cytofluorometry as-
says) revealed that Leishmania cell death was associated with
extensive nuclear DNA fragmentation. Cell-permeable
caspase inhibitors did not modify NO-mediated nuclear DNA
* Corresponding author. Mailing address: UR 008 Pathogénie des
Trypanosomatidés, IRD (Institut de Recherche pour le Développe-
ment), 911 Ave. Agropolis, BP 5045, 34032 Montpellier Cedex 1,
France. Fax: 33(0)4-67-41-63-30. Phone: 33(0)4-67-41-62-20. E-mail:
lemesre@melusine.mpl.ird.fr.
3727
fragmentation. In contrast, endonuclease activation could be a
consequence of Ca
2
-sensitive calpain and/or proteasome ac-
tivity. These data strongly suggest that NO can induce damage
in the unicellular pathogen Leishmania by a coordinated pro-
cess of intracellular protein degradation mediated by the pro-
teasome, which may lead to nuclear DNA fragmentation. An
apoptosis-like cell death pathway could represent an important
and highly regulated mechanism used for the clearance of
Leishmania within infected macrophages stimulated to pro-
duce NO endogenously or during treatments with NO-releas-
ing drugs.
MATERIALS AND METHODS
Reagents. Fetal calf serum (FCS) was obtained from Dutscher S.A. RPMI
1640 medium (lot 0MB0174) was purchased from BioWhittaker Europe, and
L-glutamine (lot 3414) was from BioMedia. Penicillin-streptomycin (10,000
IU/ml to 10,000 UG/ml; lot 3037222) and phosphate buffer saline (Ca
2
and
Mg
2
free) were obtained from Life Technologies. Bacterial lipopolysaccharide
(LPS), gamma interferon (IFN-; mouse recombinant), bovine serum albumin
(BSA; fraction V, lot 59H0696), and MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-di-
phenyltetrazolium bromide; lot 66H5033) were purchased from Sigma. RNase
was obtained from Eurogentec, proteinase K was from Promega, and agarose
was purchased from Eurobio. YOPRO-1 iodide was provided by Molecular
Probes. Nitro-L-arginine, inhibitory peptides Z-DEVD-CMK and Z-VAD-FMK,
E-64d [(2S,3S)-trans-epoxysuccinyl-L-leucylamido-3-methylbutane ethyl ester],
calpain inhibitor I, lactacystin, proteasome inhibitor II, and MG-115 were pur-
chased from Alexis Biochemicals. All other cellular-grade and molecular biolo-
gy-grade chemicals were obtained from Sigma. Cell-free MAA/20 culture me-
dium for axenically grown amastigotes was dened according to the method of
Lemesre et al. (29, 30). Nitrosylated albumin was prepared as previously de-
scribed (38).
Animals. Female BALB/c mice (Iffa Credo, Saint-Germain-sur-lArbresle,
France) were housed under conventional conditions and given water and chow
ad libitum. The use of the animals conformed to institutional guidelines.
Parasites and in vitro cultures. A cloned line of L. amazonensis (MHOM/BR/
76/LTB-012) was used in all experiments. L. amazonensis axenically grown amas-
tigote forms were maintained at 32 1°C by weekly subpassages in MAA/20
medium. From a starting inoculum of 5 10
5
amastigote forms/ml, a cell density
of ca. 2 10
7
parasites/ml was obtained on day 7. Axenically grown amastigote
forms appeared homogeneous, round to ovoid, without apparent agella, and
nonmotile. Axenically grown amastigotes from various Leishmania species
clearly resembled intracellular amastigotes with regard to their ultrastructural,
biological, biochemical, and immunological properties (5, 6, 13, 24, 29, 30, 53).
Cell concentrations were determined by daily counts with a hemocytometer at a
400 magnication after adequate dilution in phosphate-buffered saline (PBS)
containing 0.25% glutaraldehyde.
Treatment with the NO donors sodium nitrite (NaNO
2
) and nitrosylated
albumin (NO-BSA). Late-log-phase amastigote forms were washed three times
in PBS0.01 M (pH 4.5) before treatment. Parasites (10
7
/ml) were resuspended
in the same buffer containing 5 mM sodium nitrite. After 20 min, 2 h, 4 h, 6 h,
and 12 h of incubation at 32 1°C in the dark, respectively, parasites were
washed in Ca
2
-Mg
2
-free PBS (0.01 M, pH 7.2) before analysis. Control amas-
tigotes were treated in the same way but in the absence of sodium nitrite.
Albumin or nitrosylated albumin (4 mg/ml) was added to the parasite cultures.
After incubation for 20 min, 4 h, or 12 h at 32 1°C, the amastigotes were
washed three times in Ca
2
-Mg
2
-free PBS (0.01 M, pH 7.2) before analysis.
Assessment of NO donorseffect on amastigote survival. NO-treated and
untreated amastigotes (2 10
8
/ml) were resuspended in MAA/20 medium for
each incubation time. The MTT-based microassay was used to estimate NO-
induced death as described before (53). Tetrazolium bromide has the property of
being reduced by parasitic dehydrogenases into a dark blue insoluble formazan
product, the amount of which depends on the number of viable amastigotes.
Briey, parasites (100 l) were seeded in a 96-well microplate. After a 1-h
incubation, 10 l of MTT (10 mg/ml) was added to each well, and the plates were
further incubated for 4 h. The enzyme reaction was then stopped by the addition
of 100 l of 50% isopropanol10% sodium dodecyl sulfate. The plates were
incubated for an additional 30 min while being agitated at room temperature
before the optical density value at 570 nm was determined with a Titer-Tech
96-well scanner (Labsystems Multiscan EX).
In vitro macrophage infections. Mouse peritoneal macrophages were washed
with prewarmed RPMI 1640 medium supplemented with 10% heat-inactivated
FCS, 2 mM glutamine, 100 U of penicillin/ml, and 100 g of streptomycin/ml and
cultured overnight in 16-well Lab-Tek tissue culture slides (Nalge Nunc Inter-
national). Nonadherent cells were removed by two washes with prewarmed
RPMI medium, and then the macrophages were infected with stationary-phase
extracellular amastigotes at a parasite/macrophage ratio of 3:1 for2hat34°C
with 5% CO
2
. Noninternalized parasites were removed by gentle washing. In-
fected macrophages were then cultured in the presence or absence of the fol-
lowing reagents: LPS (10 ng/ml), gamma interferon (IFN-; 100 U/ml), and a
combination of LPS and IFN-. Controls were made by adding 1 mM nitro-L-
arginine, a competitive inhibitor of NOS II, or 1 mM nitro-L-arginine plus 2 mM
L-arginine to revert the inhibition.
Culture supernatants were collected for nitrite production measurement 48 h
later. Macrophages were then washed with prewarmed RPMI, xed with meth-
anol, and stained with Giemsa for parasite counts or processed for the TUNEL
technique (see below). The percent parasite index (PI) inhibition compared to
the untreated controls was calculated as follows: percent PI 100 [(mean
number of amastigotes per macrophage percent infected macrophages in
treated wells)/(mean number of amastigotes per macrophage percentage of
infected macrophages in untreated wells)] 100. The results were taken as the
mean of duplicate experiments.
Measurement of nitrite production. NO
2
accumulation in the medium over
48 h for the leishmanicidal assay was used as an indicator of NO production and
was assayed by the Griess reaction (50). Briey, 60 l of Griess reagent A (1%
sulfanilamide in 1.2 N HCl) and 60 l of Griess reagent B [0.3% N-(1-naphth-
yl)ethylenediamine] were added to 100 l of each supernatant in triplicate wells
in a 96-well plate. Plates were read at 540 nm in an enzyme-linked immunosor-
bent assay plate reader (Labsystems Multiskan EX). NaNO
2
in RPMI was used
to construct a standard curve for each plate reading.
In situ TUNEL assay. Slides with infected macrophages or axenic amastigotes
were xed for 20 min with PBS containing 4% paraformaldehyde, washed twice
with 0.01 M PBS (pH 7.2), and stored at 20°C until use. DNA fragmentation
was analyzed in situ by using a colorimetric apoptosis detection system (Promega,
Madison, Wis.) according to the manufacturers instructions. After performance
of the TUNEL protocol, preparations were analyzed under a microscope at
1,000 magnication. Apoptotic nuclei appeared dark brown. Other nuclei were
not colored.
DNA agarose gel electrophoresis. Cell pellets were incubated in lysis buffer (10
mM Tris, 10 mM EDTA, 0.5% Triton X-100 [pH 7.4]) for 30 min at 4°C.
Qualitative analysis of DNA fragmentation was performed as previously de-
scribed (7) by agarose gel electrophoresis of DNA extracted from 5 10
8
axenically grown amastigotes. DNA was then visualized under UV light after the
gels were stained with ethidium bromide.
Flow cytouorometry analysis with YOPRO-1. The percentage of apoptotic
cells was quantitated by ow cytouorometric analysis by using the DNA inter-
calatant YOPRO-1 as previously described (25). Briey, 10
6
L. amazonensis
amastigotes were incubated with 10 M YOPRO-1 for 10 min. Cells were
immediately analyzed with a FACScan ow cytometer (Becton Dickinson, Ivry,
France) by using an argon-ion laser tuned to 488 nm. Green cell uorescence,
gated on forward scatter and side light scatter, was collected by using a (525
10)-nm band-pass lter (FL1) and displayed by using a logarithmic amplication.
Statistical analysis. Statistical signicance was analyzed by Studentsttest. All
experiments were performed at least twice.
RESULTS
Kinetics of NO-mediated effect on L. amazonensis amasti-
gote viability. Treatment of L. amazonensis extracellular amas-
tigotes with both NO donors led to a dramatic loss of parasite
viability with regard to morphological changes such as de-
creased cell volume under microscopic examination (data not
shown), as well as a decrease in dehydrogenase activities as
determined by the MTT micromethod (Fig. 1). Exposure of
axenically grown amastigotes to NO generated from acidied 5
mM NaNO
2
resulted in a time-dependent cell death (Fig. 1A).
Signicant toxicity could be detected as early as 20 min after
treatment with the chemical (ca. 40% of amastigote mortality).
Survival gradually decreased; 10% of the microorganisms
were viable after a 12-h exposure to NO generated from
3728 HOLZMULLER ET AL. INFECT.IMMUN.
NaNO
2.
Incubation of amastigotes in PBS (pH 4.5) in the
absence of added NaNO
2
did not signicantly affect parasite
viability in the rst few hours, and fewer than 25% amastigotes
were dead after 12 h (Fig. 1A).
About 30% of amastigotes exposed to 4 mg of NO-BSA/ml
were killed within the rst 20 min of incubation, reaching ca. 65
and 90% after 4 and 12 h of incubation, respectively (Fig. 1B).
The addition of albumin alone at 4 mg/ml failed to affect
amastigote viability, even after 12 h of incubation (Fig. 1B),
and no striking morphological change was observed under the
microscope (not shown).
The relevance of the observations above would remain ques-
tionable unless similar effects could be observed in intracellu-
lar amastigotes of L. amazonensis exposed to endogenous NO
produced by activated macrophages. Mouse peritoneal macro-
phages infected with axenically cultured amastigotes and acti-
vated for 24 and 48 h with IFN-and LPS led to a time-
dependent intracellular killing of Leishmania as determined by
the reduction of PIs of ca. 55% at 24 h and 90% at 48 h (Fig.
2B), concomitant with NO
2
accumulation in culture uids
(Fig. 2A). NO
2
production and antileishmanial activity both
decreased in the presence of 1 mM nitro-L-arginine, a compet-
itive inhibitor of NOS II. They were almost totally restored by
the addition of 2 mM L-arginine (Fig. 2). Inhibition of the PI
did not exceed 20% when incubation conditions did not induce
NO production (Fig. 2B).
NO-mediated apoptosis-like DNA fragmentation of L. ama-
zonensis amastigotes. NO-mediated DNA fragmentation ex-
hibiting features of apoptosis was rst assessed by using extra-
cellular amastigotes incubated 5 mM NaNO
2
and monitoring
the genomic DNA status of treated versus untreated parasites.
As shown in Fig. 3 (lanes 5, 7, and 9), nuclear DNA fragmen-
tation into oligonucleosomal-sized fragments (720, 360, and
180 bp), a typical feature of apoptotic cells, was readily visible
in the case of NO-treated amastigotes. Untreated amastigotes
did not show any DNA fragmentation, even after6hofincu-
bation in acidic conditions (Fig. 3, lanes 2, 4, 6, and 8). Inter-
estingly, NO-mediated nuclear DNA fragmentation (720-bp
multimer) could be detected as early as after 2 h of contact
with the NO donor (Fig. 3, lane 5). After6hofcontact with the
chemical, the entire genomic DNA of amastigotes was mostly
fragmented into oligonucleosome-sized DNA of 360 and 180
bp (Fig. 3, lane 9), the end products of DNA alteration in
apoptosis.
Endonuclease activity was also evaluated by the TUNEL
assay. As shown in Fig. 4C, nuclei of axenically grown amasti-
gotes exposed to 5 mM NaNO
2
were intensely stained dark
FIG. 1. Effects of 5 mM acidied sodium nitrite (A) or 4 mg of
nitrosylated albumin/ml (B) on the death of L. amazonensis amasti-
gotes. Parasite viability was assessed by the MTT micromethod as
described by Sereno and Lemesre (53). Statistical signicance was
determined by the Studentsttest. Results are expressed as the mean
and standard deviation of three independent experiments made in
triplicate. Statistical signicance: ,P0.05; ⴱⴱ,P0.01; ⴱⴱⴱ,P
0.001.
FIG. 2. NO-dependent cytotoxicity against L. amazonensis in acti-
vated mouse macrophages. Macrophages were infected with L. ama-
zonensis amastigotes at a cell/parasite ratio of 1:3 in RPMI 1640 me-
dium supplemented with 10% FCS. Infected cells were treated with
LPS, IFN-, LPS plus IFN-, LPS plus IFN-plus nitro-L-arginine (a
competitive inhibitor of NOS II), and LPS plusIFN-plus nitro-L-
arginine plus L-arginine (to reverse NOS II inhibition). NO
2
levels in
supernatants (A) and PI inhibitions in cells (B) were determined 24
and 48 h later. The results represent the mean and standard deviation
of two independent experiments made in duplicate. Statistical signi-
cance: ,P0.05; ⴱⴱ,P0.01; ⴱⴱⴱ,P0.001.
VOL. 70, 2002 DNA FRAGMENTATION IN LEISHMANIA AMASTIGOTES 3729
brown after a 4-h incubation in contrast to those of untreated
parasites (Fig. 4A). Corresponding controls incubated without
terminal deoxynucleotidyl transferase (TdT) enzyme in order
to determinate the specicity of the reaction did not show any
staining (Fig. 4B and D). Interestingly, kinetoplast DNA of
NO-treated and untreated amastigotes was also stained. In-
deed, replication of kinetoplast DNA involves the release of
catenated minicircles from the network with subsequent DNA
3-OH tail production stained by biotinylated dUTP via the
TdT enzyme. Similarly, the in situ TUNEL technique revealed
that the nuclear DNA fragmentation of intracellular amasti-
gotes also occurred in Leishmania-infected stimulated macro-
phages (Fig. 5). Moreover, a strong correlation between nu-
clear DNA fragmentation, increased NO
2
levels, and
antileishmanial activity was observed (Fig. 2 and 5). Labeled
amastigote nuclei could only be visualized inside activated
macrophages producing NO (Fig. 5D and F). Nuclei of intra-
cellular amastigotes, which we can visualized after Giemsa
staining, were free of label in nonactivated macrophages (Fig.
5A) and in macrophages stimulated with LPS alone (B), with
IFN-alone (C), or with LPS and IFN-plus 1 mM nitro-L-
arginine (E). In the absence of TdT enzyme, corresponding
negative controls did not exhibit any staining, thus demonstrat-
ing the specicity of the reaction (Fig. 5G and H).
Quantitation and time course of NO-mediated cell death of
L. amazonensis amastigotes. The impermeable DNA interca-
lated YOPRO-1 has been previously used for monitoring the
apoptosis of Trypanosoma cruzi epimastigotes (3, 25). We show
here that it could also selectively differentiate viable, necrotic
(sodium dodecyl sulfate-treated), and apoptotic amastigotes
(Fig. 6). Trivalent antimonial (potassium antimonyl tartrate at
50 g/ml) was used as a positive apoptosis inducer as described
by Sereno et al. (54). Antimonial-mediated apoptotic cells
(Fig. 6C and D) were easily distinguished from living (Fig. 6E
and F) and necrotic cells (Fig. 6A and B) by using the com-
FIG. 3. DNA fragmentation analysis. Agarose gel electrophoresis of untreated amastigote (suspended in PBS [pH 4.5], lanes 2, 4, 6, and 8) and
NO-treated amastigote (suspended in PBS [pH 4.5] plus 5 mM NaNO
2
; lanes 3, 5, 7, and 9) DNA (5 g) is shown. Exposure times: 20 min (lanes
2 and 3), 2 h (lanes 4 and 5), 4 h (lanes 6 and 7), and 6 h (lanes 8 and 9); molecular weights are indicated beside lanes 1 and 10. The experiment
was done four times.
FIG. 4. In situ analysis of L. amazonensis extracellular amastigote
death exhibiting features of apoptosis. (A and B) Untreated (PBS [pH
4.5], 4 h) amastigotes (A) and, as negative control, untreated parasites
incubated without TdT (B). (C and D) NO-treated amastigotes (5 mM
NaNO
2
, 4 h) incubated with (C) or without (D) TdT. DNA fragmen-
tation analysis was determined by the TUNEL method and analyzed
under a microscope at 1,600 magnication. The experiment was
done twice.
3730 HOLZMULLER ET AL. INFECT.IMMUN.
bined analysis of their different patterns of FSC-H properties
and YOPRO-1 staining (FL1-H).
The time course of the apoptosis-like changes was deter-
mined by evaluating the percentage of cells that exhibited the
maximum uorescence seen in NO-treated amastigotes. Amas-
tigotes incubated in 0.01 M PBS (pH 4.5) for 20 min, 2 h, and
4 h (not shown) and 6 h (Fig. 6E and F) displayed a homoge-
neous population of living cells with a low uorescence back-
ground. In contrast, a new cell population with a high FL1
uorescence corresponding to apoptotic cell-like cells ap-
peared in NO-treated amastigotes. This population increased
as a function of exposure time to the NO donor, representing
ca. 14% at 20 min (not shown), 19.9% at 2 h (Fig. 6G and H),
46.9% at 4 h (Fig. 6I and J), and 88.9% at 6 h (Fig. 6K and L).
Interestingly, the time courses of YOPRO-1 staining and of
viability loss were quite similar (Fig. 1 and 6). Altogether, these
data indicated that NO directly and quickly induced extracel-
lular amastigote death through a process that mimics apopto-
sis.
Effects of apoptosis inhibitors. In higher eukaryote cells,
apoptosis often occurs downstream of the death signal by pro-
tease activation, leading to endonuclease ignition (21). In these
instances, cysteine proteases of the caspase family, Ca
2
-sen-
sitive calpains, or proteasomes can initiate cell death. We used
here cytouorometric analysis with YOPRO-1 staining to test
the relevance of these pathways with apoptosis inhibitors.
Amastigote treatment with apoptosis-blocking agents did not
alter the viability of untreated amastigotes during the time
course experiments (not shown). Whereas caspase inhibitors
reversed the apoptosis of murine peritoneal macrophages in-
duced by 4 mM butyrate as recently described (47), inhibitors
of caspase 3 (inhibitory peptide Z-DEVD-CMK at 5 M),
caspase 1 (inhibitory peptide Z-VAD-FMK at 5 M), or cys-
teine proteases (E-64d at 20 M] had no effect on the cell
death induced by NO (data not shown). In contrast, a signi-
cant time-dependent inhibitory effect was observed on the NO-
mediated apoptosis-like phenomenon with specic irreversible
proteasome inhibitors (10 M lactacystin and 20 M calpain
inhibitor I) (Fig. 7). Treatment with reversible proteasome
inhibitors (10 M proteasome inhibitor II or 10 M MG-115)
exhibited a less potent and an inversely time-dependent inhib-
itory activity. In the rst2hoftreatment, all of the proteasome
FIG. 5. In situ analysis of the apoptosis-like process of L. amazonensis amastigotes in mouse macrophages. Cells were infected with amastigote
forms at a parasite/cell ratio of 3:1 in RPMI 1640 medium supplemented with 10% FCS. Infected cells were activated for 48 h. (A) Untreated
macrophages; (B) macrophages activated with LPS; (C) macrophages activated with IFN-; (D) macrophages activated with LPS plus IFN-;
(E) macrophages activated with LPS plus IFN-plus nitro-L-arginine (a competitive inhibitor of NO synthase type II); (F) macrophages activated
with LPS plus IFN-plus nitro-L-arginine plus L-arginine (to reverse inhibition of NOS-II). (G and H) Negative controls, either activated
macrophages (G) or nonactivated macrophages (H), were incubated without TdT. DNA fragmentation analysis was determined by the TUNEL
technique and analyzed under a microscope at 1,000 magnication (A to H). A magnication of 1,600 was used to identify labeled amastigotes
in panel I. The experiment was done twice.
VOL. 70, 2002 DNA FRAGMENTATION IN LEISHMANIA AMASTIGOTES 3731
inhibitors used exerted an inhibitory effect ranging from 20 to
40%. After a 4-h incubation with proteasome inhibitor II and
MG-115, NO-induced DNA changes were reduced by ca. 25%,
whereas lactacystin and calpain inhibitor I produced marked
inhibitions of 47 and 62%, respectively. These percentages
reached ca. 50 and 64%, respectively, after6hofincubation
(Fig. 7). All of these data support the view that the proteasome
pathway could be involved in promoting apoptosis-like changes
in NO-exposed amastigotes.
DISCUSSION
In this study we showed that the leishmanicidal effect of NO
seemed to be the consequence of the induction of a pro-
grammed cell death-like process. NO donors induced a pattern
of DNA fragmentation typical of an apoptotic process in ax-
enic amastigotes of L. amazonensis. This apoptotic phenome-
non was conrmed by both the TUNEL technique and ow
cytometry. This typical aspect of apoptosis was also demon-
strated in the Leishmania intracellular parasite stage inside
NO-producing macrophages, by using the combination IFN-
and LPS, which is mainly used as a potent inducer of NOS II.
NO also seems to be able to induce necrosis, as well as
apoptosis (34). Interestingly, NO-mediated apoptosis has been
extensively studied in mammalian cells (1, 2, 12, 18, 28, 36, 63).
Recent reports have shown that unicellular organisms may kill
themselves by an evolutionarily conserved programmed cell
death pathway (i.e., apoptosis) (4). This pathway has been
described in several lower organisms, including the slime mold
Dictyostelium discoideum (11) and several parasitic protozoa.
The effect of chloroquine on a sensitive strain of the malaria
parasite P. falciparum led to an oligonucleosomal DNA frag-
mentation, suggesting that apoptosis may be involved in the
action of chloroquine on the parasite (45). Trypanosoma brucei
brucei and Trypanosoma brucei rhodesiense procyclic forms,
upon treatment with the lectin concanavalin A, died by a pro-
cess similar to apoptosis in mammals (61, 62); changes in
cytoskeletal organization, cytosol vacuolization, and nuclear
DNA fragmentation were reported. Apoptosis-like changes
have also been reported for T. cruzi in response to conditioned
medium or antibiotic G418 (3). A shift in the distribution of
EF-1to a nuclear localization was reported as one of the
changes associated with cell death (8). In leishmaniasis, a heat
FIG. 6. Analysis of the NO-induced apoptosis-like process in extracellular amastigotes by a cytouorometry YOPRO-1 differential staining
technique. Parasites were incubated for6hin0.01 M PBS (pH 7.2) (negative control, not shown), treated for 10 min by 0.1% sodium dodecyl
sulfate (necrosis control, A and B), and treated for 24 h with 50 g of potassium antimonyl tartrate/ml (apoptosis control, C and D). Amastigotes
were, respectively, incubated in 0.01 M PBS (pH 4.5) as an internal control (a 6-h incubation is depicted in E and F) and treated with 5 mM NaNO
2
for2h(GandH),4h(IandJ),or6h(KandL).Theapoptotic cell percentage (R2), corresponding to both reduced forward scatter and high
uorescence intensity, is indicated in each experimental condition. M1 shows the peak of uorescence intensity in panels B, D, F, H, J, and L. The
experiment was done three times in duplicate.
3732 HOLZMULLER ET AL. INFECT.IMMUN.
shock was able to induce in promastigotes of L. amazonensis a
DNA cleavage into an oligonucleosomal ladder characteristic
of cells dying by apoptosis (39). In the same way, luteolin and
quercetin, two plant-derived avonoids, inhibited cell cycle
progression in L. donovani promastigotes, leading to apoptosis
(37). Recently, we demonstrated that the antileishmanial tox-
icity of trivalent antimonials is associated with parasite oligo-
nucleosomal DNA fragmentation, a nding indicative of the
occurrence of late events in the overall apoptosis process (54).
However, the activation of an apoptosis-like machinery in the
parasite stage directly in contact with macrophage antimicro-
bial agents such as NO (i.e., amastigotes) has never been in-
vestigated. In the present study, we demonstrated that both
axenically grown and intracellular L. amazonensis amastigotes,
when submitted to NO action, commit cell suicide through a
pathway that is referred to as apoptosis.
In the last few years it has been established that protease
activationin cysteine proteases of the caspase family in par-
ticularis an integral component of the apoptotic cell death
metabolic pathway (64). We report here that cell-permeable
caspase inhibitors had no effect on the oligonucleosomal DNA
fragmentation induced by NO on L. amazonensis amastigotes.
This result correlated with observations made on T. brucei
brucei exposed to reactive oxygen species (48). The inhibitors
used are known, at least, to block caspases that are directly and
indirectly responsible for the activation of endonuclease activ-
ity (i.e., caspases 3 and 1, respectively). NOS II induction is
triggered and regulated by a series of signaling pathways in-
ducing NF-B (56). Inhibitors of the proteasome and caspase
3 abrogate the induction of NOS II in macrophages by block-
ing activation of the transcription factor NF-B (9, 23). These
inhibitors cannot be used in culture models relying on NOS II
induction. Thus, our experiments of inhibition were restricted
to axenically grown amastigotes. Surprisingly and in contrast to
data obtained from African trypanosomes by Ridgley et al.
(48), specic irreversible proteasome inhibitors (lactacystin
and calpain inhibitor I) markedly inhibited the NO-induced
oligonucleosomal DNA fragmentation of Leishmania extracel-
lular amastigotes, suggesting that an NO-mediated apoptosis-
like process needed proteasome activity to work. This diver-
gence with the observations made by Ridgley et al. (48) might
be explained by the status of cell maturation of the parasite
considered (35). Moreover, evidence is now accumulating that
noncaspases, including cathepsins, calpains, granzymes, and
the proteasome complex, also have roles in mediating and
promoting cell death (27, 43). Recent studies have otherwise
pointed out in two Leishmania species the existence of an
active proteasome, one similar to the proteasomes of other
eukaryotes (10, 49). This potent proteasome might be involved
in the NO-mediated apoptosis-like process. This nding sug-
gests the existence of an ancestral noncaspase protease cell
death pathway.
Finally, NO-mediated cytostatic and cytotoxic effects on ax-
enically grown amastigotes were thus a consequence of the
activation of a cell death program exhibiting features of apo-
ptosis. A proteasome-dependent apoptosis-like machinery is
also present and active in the clinically relevant stage of the
parasite (i.e., amastigote) and can be induced in the host cell by
the L-arginineNO pathway. However, this apoptotic event
might limit the hostsinammatory and specic immune re-
sponses, leading to a decrease in NO-dependent parasite kill-
ing and favoring the persistence of some parasites. Neverthe-
less, NO donors have been used to cure murine and human
cutaneous leishmaniasis (15, 65). The investigation of an apo-
ptotic process in cells from NO-treated lesions might merit
further research.
ACKNOWLEDGMENT
We thank Phil Agnew for reviewing the manuscript and for many
useful suggestions.
REFERENCES
1. Albina, J. E., and J. S. Reichner. 1998. Role of nitric oxide in mediation of
macrophage cytotoxicity and apoptosis. Cancer Metastasis Rev. 17:3953.
2. Albina, J. E., S. Cui, R. B. Mateo, and J. S. Reichner. 1993. Nitric oxide-
mediated apoptosis in murine peritoneal macrophages. J. Immunol. 150:
50805085.
3. Ameisen, J. C., T. Idzioreck, O. Billaut-Mulot, M. Loyens, J. P. Tissier, A.
Potentier, and A. Ouaissi. 1995. Apoptosis in a unicellular eukaryote
(Trypanosoma cruzi): implications for the evolutionary origin and the control
of programmed cell death in the control of cell proliferation, differentiation
and survival. Cell. Death Differ. 2:285300.
4. Ameisen, J. C. 1996. The origin of programmed cell death. Science 272:1278
1279.
5. Bates, P. A. 1993. Characterization of developmentally regulated nucleases
in promastigotes and amastigotes of Leishmania mexicana. FEMS Microbiol.
Lett. 107:5358.
6. Bates, P. A., C. D. Robertson, L. Tetley, and G. H. Coombs. 1992. Axenic
cultivation and characterization of Leishmania mexicana amastigote-like
forms. Parasitology 105:193202.
7. Berman, J. D., J. V. Gallalee, and J. M. Best. 1987. Sodium stibogluconate
(Pentostam) inhibition of glucose catabolism via the glycolytic pathway, and
fatty acid beta-oxidation in Leishmania mexicana amastigotes. Biochem.
Pharmacol. 36:197201.
8. Billaut-Mulot, O., R. Fernandez-Gomez, M. Loyens, and A. Ouaissi. 1996.
Trypanosoma cruzi elongation factor 1-alpha: nuclear localization in para-
sites undergoing apoptosis. Gene 174:1926.
9. Chakravortty, D., Y. Kato, T. Sugiyama, N. Koide, M. M. Mu, T. Yoshida,
and T. Yokoshi. 2001. Inhibition of caspase-3 abrogates lipopolysaccharide-
induced nitric oxide production by preventing activation of NF-B and c-jun
NH
2
-terminal kinase/stress-activated protein kinase in RAW 264.7 murine
macrophage cells. Infect. Immun. 69:13151321.
10. Christensen, C. B., L. Jorgensen, A. T. Jensen, S. Gasim, M. Chen, A.
Kharazmi, T. G. Theander, and K. Andresen. 2000. Molecular characteriza-
tion of a Leishmania donovani cDNA clone with similarity to human 20S
proteasome a-type subunit. Biochim. Biophys. Acta 1500:7787.
FIG. 7. Percentage of apoptotic cells, in the presence of protea-
some inhibitors, as measured by ow cytometry and YOPRO-1 stain-
ing of NO-treated amastigotes of L. amazonensis (5 mM NaNO
2
).
Incubation media were supplemented with the following proteasome
inhibitors: lactacystin (10 M), calpain inhibitor I (20 M), protea-
some inhibitor II (10 M), and MG-115 (10 M). Statistical signi-
cance was determined by the Studentsttest. The results are the means
the standard deviation of three independent experiments made in
duplicate. Statistical signicance: ,P0.05; ⴱⴱ,P0.01; ⴱⴱⴱ,P
0.001.
VOL. 70, 2002 DNA FRAGMENTATION IN LEISHMANIA AMASTIGOTES 3733
11. Cornillon, S., C. Foa, J. Davoust, N. Buonavista, J. D. Gross, and P. Gol-
stein. 1994. Programmed cell death in Dictyostelium. J. Cell Sci. 107:2691
2704.
12. Cui, S., J. S. Reichner, R. B. Mateo, and J. E. Albina. 1994. Activated murine
macrophages induce apoptosis in tumor cells through nitric oxide-dependent
or -independent mechanisms. Cancer Res. 54:24622467.
13. Cysne-Finkelstein, L., R. M. Temporal, F. A. Alves, and L. L. Leon. 1998.
Leishmania amazonensis: long-term cultivation of axenic amastigotes is as-
sociated to metacyclogenesis of promastigotes. Exp. Parasitol. 89:5862.
14. Das, L., N. Datta, S. Bandyopadhyay, and P. K. Das. 2001. Successful
therapy of lethal murine visceral leishmaniasis with cystatin involves upregu-
lation of nitric oxide and a favorable T-cell response. J. Immunol. 166:4020
4028.
15. Davidson, R. N., V. Yardley, S. L. Croft, P. Konecny, and N. Benjamin. 2000.
A topical nitric oxide-generating therapy for cutaneous leishmaniasis. Trans.
R. Soc. Trop. Med. Hyg. 94:319322.
16. Duhe, R. J., M. D. Nielsen, A. H. Dittman, E. C. Villacres, E. J. Choi, and
D. R. Storm. 1994. Oxidation of critical cysteine residues of type I adenyl
cyclase by o-iodosobenzoate or nitric oxide reversibly inhibits stimulation by
calcium and calmodulin. J. Biol. Chem. 269:72907296.
17. Evans, T. G., L. Thai, D. L. Granger, and J. B., Jr. Hibbs. 1993. Effect of in
vivo inhibition of nitric oxide production in murine leishmaniasis. J. Immu-
nol. 151:907915.
18. Fortenberry, J. D., M. L. Owens, M. R. Brown, D. Atkinson, and L. A. S.
Brown. 1998. Exogenous nitric oxide enhance neutrophil cell death and
DNA fragmentation. Ann. J. Respir. Cell Mol. Biol. 18:421428.
19. Girard, P., and P. Potier. 1993. NO, thiols, and disuldes. FEBS Lett.
320:78.
20. Gobert, A. P., S. Semballa, S. Daulouede, S. Lesthelle, M. Taxile, B. Veyret,
and P. Vincendeau. 1998. Murine macrophages use oxygen- and nitric oxide-
dependent mechanisms to synthesize S-nitroso-albumin and to kill extracel-
lular trypanosomes. Infect. Immun. 66:40684072.
21. Green, D., and G. Kroemer. 1998. The central executioners of apoptosis:
caspases or mitochondria? Trends Cell. Biol.8:267271.
22. Green, S. J., M. S. Meltzer, J. B. Hibbs, Jr., and C. A. Nacy. 1990. Activated
macrophages destroy intracellular Leishmania major amastigotes by an L-
arginine-dependent killing mechanism. J. Immunol. 144:278283.
23. Griscavage, J. M., S. Wilk, and L. J. Ignarro. 1996. Inhibitors of the pro-
teasome pathway interfere with induction of nitric oxide synthase in macro-
phages by blocking activation of transcription factor NF-B. Proc. Natl.
Acad. Sci. USA 93:33083312.
24. Hodgkinson, V. H., L. Soong, S. M. Duboise, and D. McMahon-Pratt. 1996.
Leishmania amazonensis: cultivation and characterization of axenic amasti-
gote-like organisms. Exp. Parasitol. 83:94105.
25. Idziorek, T., J. Estaquier, F. De Bels, and J. C. Ameisen. 1995. YOPRO-1
permits cytouorometric analysis of programmed cell death (apoptosis)
without interfering with cell viability. J. Immunol. Methods 185:249258.
26. James, S. L., and C. Nacy. 1993. Effector functions of activated macrophages
against parasites. Curr. Opin. Immunol. 5:518523.
27. Johnson, D. E. 2000. Noncaspase proteases in apoptosis. Leukemia 14:1695
1703.
28. Lancaster, J. R. 1996. Diffusion of free nitric oxide. Methods Enzymol.
268:3150.
29. Lemesre, J. L., D. Sereno, S. Daulouède, B. Veyret, N. Brajon, and P.
Vincendeau. 1997. Leishmania spp.: nitric oxide-mediated metabolic inhibi-
tion of promastigote and axenically grown amastigote forms. Exp. Parasitol.
86:5868.
30. Lemesre, J. L., M. P. Blanc, P. Grebaut, V. Zilberfarb, and V. Carrière. 1994.
Culture continue des formes amastigotes de leishmanies en condition
axénique: réalisation du cycle évolutif in vitro. Med. Armee 22:99100.
31. Lepoivre, M., J. M. Flaman, P. Bobe, G. Lemaire, and Y. Henry. 1994.
Quenching of the tyrosyl free radical of ribonucleotide reductase by nitric
oxide: relationship to cytostasis induced in tumor cells by cytotoxic macro-
phages. J. Biol. Chem. 269:2189121897.
32. Mauel, J., and A. Ransijn. 1997. Leishmania spp.: mechanisms of toxicity of
nitrogen oxidation products. Exp. Parasitol. 87:98111.
33. Mauel, J., A. Ransijn, and Y. Buchmuller-Rouiller. 1991. Killing of Leish-
mania parasites in activated murine macrophages is based on an L-arginine-
dependent process that produces nitrogen derivatives. J. Leukoc. Biol. 49:
7382.
34. Melino, G., F. Bernassola, R. A. Knight, M. T. Corasaniti, G. Nistico, and A.
Finazzi-Agro. 1997. S-nitrosylation regulates apoptosis. Nature 388:432433.
35. Meriin, A. B., V. L. Gabai, J. Yaglom, V. I. Shifrin, and M. Y. Sherman. 1998.
Proteasome inhibitors activate stress kinases and induce Hsp72: diverse
effects on apoptosis. J. Biol. Chem. 273:63736379.
36. Messmer, U. K., U. K. Reed, and B. Brune. 1996. Bcl-2 protects macrophages
from nitric oxide-induced apoptosis. J. Biol. Chem. 271:2019220197.
37. Mittra, B., A. Saha, A. R. Chowdhury, C. Pal, S. Mandal, S. Mukhopadhyay,
S. Bandyopadhyay, and H. K. Majumder. 2000. Luteolin, an abundant di-
etary component is a potent antileishmanial agent that acts by inducing
topoisomerase II-mediated kinetoplast DNA cleavage leading to apoptosis.
Mol. Med. 6:527541.
38. Mnaimneh, S., M. Geffard, B. Veyret, and P. Vincendeau. 1997. Albumin
nitrosylated by activated macrophages possesses antiparasitic effects neu-
tralized by anti-NO-acetylated-cysteine antibodies. J. Immunol. 158:308
314.
39. Moreira, M. E., H. A. Del Portillo, R. V. Milder, J. M. Balanco, and M. A.
Barcinski. 1996. Heat shock induction of apoptosis in promastigotes of the
unicellular organism Leishmania (Leishmania)amazonensis. J. Cell Physiol.
167:305313.
40. Mossalayi, M. D., M. Arock, D. Mazier, P. Vincendeau, and I. Vouldoukis.
1999. The human immune response during cutaneous leishmaniasis: NO
problem. Parasitol. Today 15:342345.
41. Muhl, H., K. Sandau, B. Brune, V. A. Briner, and J. Pfeilschifter. 1996.
Nitric oxide donors induce apoptosis in glomerular mesangial cells, epithelial
cells and endothelial cells. Eur. J. Pharmacol. 317:137149.
42. Nomura, Y., T. Uehara, and M. Nakazawa. 1996. Neuronal apoptosis by glial
NO: involvement of inhibition of glyceraldehyde-3-phosphate dehydroge-
nase. Hum. Cell 9:205214.
43. Orlowski, R. Z. 1999. The role of the ubiquitin-proteasome pathway in
apoptosis. Cell. Death Differ. 6:303313.
44. Panaro, M. A., A. Acquafredda, S. Lisi, D. D. Lofrumento, T. Trotta, R.
Satalino, V. Mitolo, and O. Brandonisio. 1999. Inducible nitric oxide syn-
thase and nitric oxide production in Leishmania infantum-infected human
macrophages stimulated with interferon-gamma and bacterial lipopolysac-
charide. Int. J. Clin. Lab. Res. 29:122127.
45. Picot, S., J. Burnod, V. Bracchi, B. F. Chumpitazi, and P. Ambroise-
Thomas. 1997. Apoptosis related to chloroquine sensitivity of the human
malaria parasite Plasmodium falciparum. Trans. R. Soc. Trop. Med. Hyg.
91:590591.
46. Pinelli, E., D. Gebhard, A. M. Mommaas, M. van Hoeij, J. A. Langermans,
E. J. Ruitenberg, and V. P. Rutten. 2000. Infection of a canine macrophage
cell line with Leishmania infantum: determination of nitric oxide production
and antileishmanial activity. Vet. Parasitol. 92:181189.
47. Ramos, M. G., F. L. A. Rabelo, T. Duarte, R. T. Gazzinelli, and J. I.
Alvarez-Leite. 2002. Butyrate induces apoptosis in murine macrophages via
caspase-3, but independent of autochrine synthesis of tumor necrosis factor
and nitric oxide. Braz. J. Med. Biol. Res. 35:161173.
48. Ridgley, E. L., Z. H. Xiong, and L. Ruben. 1999. Reactive oxygen species
activate a Ca
2
-dependent cell death pathway in the unicellular organism
Trypanosoma brucei brucei. Biochem. J. 340:3340.
49. Robertson, C. D. 1999. The Leishmania mexicana proteasome. Mol. Bio-
chem. Parasitol. 103:4960.
50. Roy, J. B., and R. G. Wilkerson. 1984. Fallibility of Griess (nitrite) test.
Urology 23:270271.
51. Salvati, L., M. Mattu, M. Colasanti, A. Scalone, G. Venturini, L. Gradoni,
and P. Ascenzi. 2001. NO donors inhibit Leishmania infantum cysteine pro-
teinase activity. Biochim. Biophys. Acta 1545:357366.
52. Scott, P. 1991. IFN-modulates the early development of Th1 and Th2
responses in a murine model of cutaneous leishmaniasis. J. Immunol. 147:
31493155.
53. Sereno, D., and J. L. Lemesre. 1997. Axenically cultured amastigote forms as
an in vitro model for investigation of antileishmanial agents. Antimicrob.
Agents Chemother. 41:972976.
54. Sereno, D., P. Holzmuller, I. Mangot, G. Cuny, A. Ouaissi, J. L. Lemesre.
2001. Antimonial-mediated DNA fragmentation in Leishmania infantum
amastigotes. Antimicrob. Agents Chemother. 45:20642069.
55. Sisto, M., O. Brandonisio, M. A. Panaro, M. A., A. Acquafredda, A., D.
Leogrande, A. Fasanella, T. Trotta, L. Fumarola, and V. Mitolo. 2001.
Indicible nitric oxide synthase expression in Leishmania-infected dog mac-
rophages. Comp. Immunol. Microbiol. Infect. 24:247254.
56. Tsai, S. H., S. Y. Lin-Shiau, and J. K. Lin. 1999. Suppression of nitric oxide
synthase and the downregulation of the activation of NFB in macrophages
by resveratrol. Br. J. Pharmacol. 126:673680.
57. Vincendeau, P., S. Daulouede, B. Veyret, M. L. Darde, B. Bouteille, and
J. L. Lemesre. 1992. Nitric oxide-mediated cytostatic activity on Trypano-
soma brucei gambiense and Trypanosoma brucei brucei. Exp. Parasitol.
75:353360.
58. Vouldoukis, I., P. A. Becherel, V. Riveros-Moreno, M. Arock, O. da Silva, P.
Debre, D. Mazier, and M. D. Mossalayi. 1997. Interleukin-10 and interleu-
kin-4 inhibit intracellular killing of Leishmania infantum and Leishmania
major by human macrophages by decreasing nitric oxide generation. Eur.
J. Immunol. 27:860865.
59. Vouldoukis, I., J. C. Drapier, A. K. Nussler, Y. Tselentis, O. A. Da Silva, M.
Gentilini, D. M. Mossalayi, L. Monjour, and B. Dugas. 1996. Canine visceral
leishmaniasis: successful chemotherapy induces macrophage antileishmanial
activity via the L-arginine nitric oxide pathway. Antimicrob. Agents Che-
mother. 40:253256.
60. Vouldoukis, I., V. Riveros-Moreno, B. Dugas, F. Ouaaz, P. Becherel, P.
Debre, S. Moncada, and M. D. Mossalayi. 1995. The killing of Leishmania
major by human macrophages is mediated by nitric oxide induced after
ligation of the Fc epsilon RII/CD23 surface antigen. Proc. Natl. Acad. Sci.
USA 92:78047808.
61. Welburn, S. C., and N. B. Murphy. 1998. Prohibitin and RACK homologues
3734 HOLZMULLER ET AL. INFECT.IMMUN.
are upregulated in trypanosomes induced to undergo apoptosis and in nat-
urally occurring terminally differentiated forms. Cell. Death Differ. 5:615
622.
62. Welburn, S. C., S. Lillico, and N. B. Murphy. 1999. Programmed cell death
in procyclic form Trypanosoma brucei rhodesiense: identication of differen-
tially expressed genes during Con A-induced death. Mem. Inst. Oswaldo
Cruz 94:229234.
63. Xie, K., Y. Wang, S. Huang, L. Xu, D. Bielenberg, T. Salas, D. J. McConkey, W.
Jiang, and I. J. Fidler. 1997. Nitric oxide-mediated apoptosis of K-1735 mela-
noma cells is associated with downregulation of Bcl-2. Oncogene 15:771779.
64. Yuan, J., S. Shaham, S. Ledoux, H. M. Ellis, and H. R. Horvitz. 1993. The
C. elegans cell death gene ced-3 encodes a protein similar to mammalian
interleukin-1-converting enzyme. Cell 75:641652.
65. Zeina, B., C. Baneld, and S. Al-Assad. 1997. Topical glyceryl trinitrate: a
possible treatment for cutaneous leishmaniasis. Clin. Exp. Dermatol. 22:244
245.
Editor: B. B. Finlay
VOL. 70, 2002 DNA FRAGMENTATION IN LEISHMANIA AMASTIGOTES 3735
... These ROS molecules trigger systemic production of nitric oxide by activating inducible NO synthase (iNOS). Nitric oxide can induce caspase-independent apoptosis in L. amazoniensis, characterized by the degradation of DNA (Holzmuller et al. 2002). The induction of diptericin, attacin, and defensins from fat bodies triggered by gut NO in response to trypanosomal infection has already been described in tsetse flies (Hao et al. 2001). ...
Article
Full-text available
Leishmaniasis transmission cycles are maintained and sustained in nature by the complex crosstalk of the Leishmania parasite, sandfly vector, and the mammalian hosts (human, as well as zoonotic reservoirs). Regardless of the vast research on human host-parasite interaction, there persists a substantial knowledge gap on the parasite’s development and modulation in the vector component. This review focuses on some of the intriguing aspects of the Leishmania-sandfly interface, beginning with the uptake of the intracellular amastigotes from an infected host to the development of the parasite within the sandfly’s alimentary canal, followed by the transmission of infective metacyclic stages to another potential host. Upon ingestion of the parasite, the sandfly hosts an intricate repertoire of immune barriers, either to evade the parasite or to ensure its homeostatic coexistence with the vector gut microbiome. Sandfly salivary polypeptides and Leishmania exosomes are co-egested with the parasite inoculum during the infected vector bite. This has been attributed to the modulation of the parasite infection and subsequent clinical manifestation in the host. While human host–based studies strive to develop effective therapeutics, a greater understanding of the vector-parasite-microbiome and human host interactions could help us to identify the targets and to develop strategies for effectively preventing the transmission of leishmaniasis.
... [40,[42][43][44][45][46] By the antimicrobial mechanisms of these secondary metabolites, studies revealed that the flavonoids, and phenolic compounds displayed their antimicrobial mechanisms of action via the suppression of nucleic acid creation, blockage of cytoplasmic membrane function, suppression of energy metabolism, prevent bacterial virulence factors, display a synergistic effect with current synthetic drugs, etc. [47] Today, NO-dependent cytostatic and cytotoxic effects induced by triggered macrophages against a number of parasites especially intracellular ones have been proven. [48][49][50][51][52] Studies showed that CL, NO-producing agents which used topically to treat lesions demonstrated a promising efficacy in mice models. [53] Furthermore, animal investigations have revealed that effective chemotherapies for visceral leishmaniasis are associated with the activation of the NO pathway. ...
Article
Full-text available
Background: In recent years, increasing resistance to synthetic agents, their long-term treatment, and lasting side effects, has faced many problems in treatment of leishmaniasis; so that finding a new high-efficacy antileishmanial drug with minimal side effects seems very necessary. This experimental study was aimed to evaluate the in vitro and in vivo leishmanicidal activity and cellular mechanisms of Astraglus spinosus methanolic extract (ASME) against Leishmania major infection. Materials and Methods: In vitro antileishmanial effect of ASME was evaluated on intracellular amastigotes of L. major in macrophage model. The effect of ASME on the NO production of macrophage cells was determined based on the Griess reaction for nitrites. Effect of ASME on the caspase-3-like activity of L. major promastigotes was performed according to the measuring the rate of color spectrophotometry. The 50% cytotoxic concentrations (CC 50) of the ASME on macrophages were measured to assess the cytotoxicity of ASME. In addition, in vivo effects of ASME were evaluated in infected BALB/c mice by measuring of the diameter of CL lesions and parasite load in the tested mice before and after 28 days of therapy. Results: The mean number of intracellular amastigotes of L. major significantly (P < 0.001) decreased with the IC 50 value of 36.9 ± 3.012 μg/mL and 44.3 ± 3.012 μg/mL for ASME and MA, respectively. Although more NO was produced by increasing the concentrations of the ASME, but, a notable rise was detected at the IC 50 (P < 0.001). ASME especially at the concentrations of ½ IC 50 and IC 50 significantly provoked the caspase-3 activation, by 10.3%, 25.6%, and 29.8%, respectively. The measured CC 50 value of ASME and MA was 463.3 μg/mL and 835 μg/mL, respectively. Treatment of the infected mice with various doses of ASME (50 and 100 mg/kg for 28 days), markedly declined the mean diameter of the CL lesions and parasite load in tested mice. Conclusion: Based on the obtained results, ASME can be considered as a promising herbal drug candidate for the isolation and production of a new alternative agent for CL treatment. As a result, this survey presented adequate results in the parasite eliminating in both in vitro and in vivo assay. Nevertheless, additional studies are needed to elucidate the accurate mechanisms of action of ASME and its effectiveness in clinical subjects.
... Nitric oxide is one of the main molecules utilised by macrophage against leishmania, which involves the macrophage undertaking the oxidative burst mechanism. This produces high quantities of nitric oxide and ROS which effectively facilitates the elimination of promastigotes within the macrophage, thereby limiting the population size within the host (69). Whilst the ROS induced mechanism of anti-parasitic activity is well understood, the same does not apply to nitric oxide as its specific mechanism is still not fully understood. ...
Article
Full-text available
Cutaneous parasites are identified by their specific cutaneous symptoms which are elicited based on the parasite’s interactions with the host. Standard anti-parasitic treatments primarily focus on the use of specific drugs to disrupt the regular function of the target parasite. In cases where secondary infections are induced by the parasite itself, antibiotics may also be used in tandem with the primary treatment to deal with the infection. Whilst drug-based treatments are highly effective, the development of resistance by bacteria and parasites, is increasingly prevalent in the modern day, thus requiring the development of non-drug based anti-parasitic strategies. Cutaneous parasites vary significantly in terms of the non-systemic methods that are required to deal with them. The main factors that need to be considered are the specifically elicited cutaneous symptoms and the relative cutaneous depth in which the parasites typically reside in. Due to the various differences in their migratory nature, certain cutaneous strategies are only viable for specific parasites, which then leads to the idea of developing an all-encompassing anti-parasitic strategy that works specifically against cutaneous parasites. The main benefit of this would be the overall time saved in regards to the period that is needed for accurate diagnosis of parasite, coupled with the prescription and application of the appropriate treatment based on the diagnosis. This review will assess the currently identified cutaneous parasites, detailing their life cycles which will allow for the identification of certain areas that could be exploited for the facilitation of cutaneous anti-parasitic treatment.
... Nowadays, it has been demonstrated that NO, which is produced by a number of immune cells, plays a critical role in the immune-mediated response for eliminating intracellular pathogens [53]. We reported that RJ, 10-H2DA, 10-HDAA, and sebacic acid (1, 2, and 4 μg/mL) significantly (P < 0.05) increased the production of NO as a dose-dependent pattern in comparison to the nontreated macrophage cells. ...
Article
Full-text available
Natural products and their derivatives as an inexpensive, accessible, and useful alternative medicine are broadly applied for the treatment of a wide range of diseases and infectious ones. The present study was designed to evaluate the insecticidal, antimalarial, antileishmanial, and cytotoxic effects of royal jelly and its three main fatty acids (trans-10-hydroxy-2-decenoic acid (10-H2DA), 10-hydroxydecanoic acid (10-HDAA), sebacic acid (1,10-decanedioic acid)). Insecticidal activity of RJ and 10-H2DA, 10-HDAA, and sebacic acid was performed against healthy 4th instar larvae at 25 ± 2°C. Antiplasmodial and antileishmanial effects of RJ and 10-H2DA, 10-HDAA, and sebacic acid were also performed against chloroquine-resistant Plasmodium falciparum K1-strain and Leishmania major amastigotes according to the Malstat method and macrophage model, respectively. In addition, the level of nitric oxide (NO) production in J774-A1 macrophages cells, plasma membrane permeability, and caspase-3-like activity and cytotoxicity effects of RJ and 10-H2DA, 10-HDAA, and sebacic acid against human embryonic kidney 293 (HEK239T cells) were evaluated. Considering the insecticidal activity, the results showed that the lethal concentration 50% value for RJ, 10-H2DA, 10-HDAA, and sebacic acid was 24.6, 31.4, 37.8, and 44.7 μg/mL μg/mL, respectively. RJ, 10-H2DA, 10-HDAA, and sebacic acid showed potent (P
... At present, NO, which is generated by various immune cells, plays a critical role in the immune-mediated response for eliminating intracellular pathogens such as Leishmani [39]. In the current work, ZSCME at the concentrations of 25 and 50 µg/mL meaningfully (p < 0.05) provoked the production and release of NO as a dose-dependent pattern in comparison with non-treated macrophage cells. ...
Article
Full-text available
This experimental investigation was designed to assess the in vitro and in vivo antileishmanial effects of Z. spina-christi methanolic extract (ZSCME) and also aims to assess some of the antileishmanial mechanisms such as the NO production, apoptosis, and plasma membrane permeability. We assessed the in vitro leishmanicidal effects of ZSCME (10–200 µg/mL) against intracellular amastigote stage of the Leishmania major (MRHO/IR/75/ER) and, then, in vivo examined male BALB/c mice infected by L. major. In addition, the rate of infectivity, Caspase 3 activity, nitric oxide (NO) production, the plasma membrane permeability, and the cytotoxic effects of ZSCME were studied. The primary phytochemical analysis of ZSCME revealed the existence of high amounts of flavonoids, tannins, glycosides, alkaloids, and saponin in this plant. The findings exhibited that ZSCME meaningfully (p < 0.001) reduced the viability of amastigotes of L. major, whereas it prompted the creation and release of NO, apoptosis, and the plasma membrane permeability (p < 0.05) and indicated no cytotoxicity in macrophage cells. The in vivo results also demonstrated that ZSCME significantly decreased the parasite load and the diameter of the lesions in the infected mice. Our results demonstrate the promising in vitro and in vivo antileishmanial effects of ZSCME against of L. major. Although the findings of the present study showed some possible antileishmanial mechanisms of ZSCME, such as stimulating NO production, apoptosis, and increasing plasma membrane permeability, additional investigations are required to confirm these results.
... Macrophages that are activated by cytokines can produce large amounts of nitric oxide, one of its functions is a defense against intracellular pathogens particularly Leishmania. Nitric oxide has been demonstrated to kill leishmania parasites by inducing amastigotes apoptosis (53). IFN-γ is important since it has immune-stimulatory, immune regulatory, and immune-modulatory effects. ...
Article
Full-text available
Background: This study was performed to evaluate in vitro and in vivo Leishmanicidal potential of morphine (Mph), imiquimod (IQ), and their combination. Methods: Leishmania infantum promastigote and amastigote assays were performed at the presence of 0.015–150µM Mph, 0.04–416µM IQ, and their combination. The inhibition effects of these drugs on promastigotes were evaluated after 24, 48, and 72h. The cytotoxic effects of the drugs were evaluated by MTT as well as flow cytometry after 72h. We explored the therapeutic effects of Mph and IQ in BALB/c mice at the end of the treatment using parasite load de­termination and cytokine assay. One group of mice received Mph for three weeks before infection. Results: The results of promastigote and amastigote assays showed the cytotoxic effects of the drugs at low concentra­tions. The cytotoxic effects were higher on promastigotes than amastigotes (p< 0.05). There was a negative correlation between drug concentration and amastigote/promastigote viability. Imiquimod alone or combined with Mph showed remarkable cytotoxic effects at all concentrations (p< 0.05). Flow cytometry results revealed apoptosis in the parasite following exposure to the drug combinations. Accordingly, the reduction of parasite loads in the spleen and liver was observed (p< 0.05) with simultaneous increases in IFN-γ and IL-4. We believe that the in vivo leishmanicidal effect was mediated by Mph through IL-4 and by IQ through both IL-4 and IFN-γ. Conclusion: Results pointed out the promising effects of Mph and IQ at low concentrations, especially when combined.
... As NO is a relatively stable uncharged molecule it can cross membranes such as the PV and the plasma membrane (75). NO mediates its cytotoxic effect by: a) reacting with heme-cytochrome C oxidase in pathogen mitochondria to initiate ROS production; b) compromising DNA synthesis by inhibition of ribonucleotide reductase activity; c) causing direct DNA damage (47,76); d) reacting with ROS forming peroxynitrite anion (ONOO-), which is a stronger oxidant that promotes pathogen killing by compromising mitochondrial respiration and mediating tyrosine nitration that impacts proteins structure and function (77). Finally, NO can inhibit overall metabolism activity in Leishmania parasites restricting their growth (78). ...
Article
Full-text available
Intracellular phagosomal pathogens represent a formidable challenge for innate immune cells, as, paradoxically, these phagocytic cells can act as both host cells that support pathogen replication and, when properly activated, are the critical cells that mediate pathogen elimination. Infection by parasites of the Leishmania genus provides an excellent model organism to investigate this complex host-pathogen interaction. In this review we focus on the dynamics of Leishmania amazonensis infection and the host innate immune response, including the impact of the adaptive immune response on phagocytic host cell recruitment and activation. L. amazonensis infection represents an important public health problem in South America where, distinct from other Leishmania parasites, it has been associated with all three clinical forms of leishmaniasis in humans: cutaneous, muco-cutaneous and visceral. Experimental observations demonstrate that most experimental mouse strains are susceptible to L. amazonensis infection, including the C57BL/6 mouse, which is resistant to other species such as Leishmania major , Leishmania braziliensis and Leishmania infantum . In general, the CD4 ⁺ T helper (Th)1/Th2 paradigm does not sufficiently explain the progressive chronic disease established by L. amazonensis , as strong cell-mediated Th1 immunity, or a lack of Th2 immunity, does not provide protection as would be predicted. Recent findings in which the balance between Th1/Th2 immunity was found to influence permissive host cell availability via recruitment of inflammatory monocytes has also added to the complexity of the Th1/Th2 paradigm. In this review we discuss the roles played by innate cells starting from parasite recognition through to priming of the adaptive immune response. We highlight the relative importance of neutrophils, monocytes, dendritic cells and resident macrophages for the establishment and progressive nature of disease following L. amazonensis infection.
Article
Background: We decided to investigate the antileishmanial, cellular mechanisms, and cytotoxic effects of green synthesized Zinc nanoparticles (ZnNPs) alone and combined with glucantime against Leishmania major infection. Methods: The effect of green synthesized ZnNP on L. major amastigote was studied through macrophage cells. The mRNA expression level of iNOS and IFN-γ followed by the exposure of J774-A1 macrophage cells to ZnNPs was assessed by Real-time PCR. The Caspase-3-like activity of promastigotes exposed to ZnNPs was studied. Effects of ZnNPs alone and combined with glucantime (MA) were studied on cutaneous leishmaniasis in BALB/c mice. Results: ZnNPs displayed the spherical shape with sizes ranging from 30 to 80 nm. The obtained IC50 values for ZnNPs, MA, and ZnNPs + MA were 43.2, 26.3, and 12.6 µg/mL, respectively; indicating the synergistic effects of ZnNPs in combination with MA. CL lesions had completely improved in the mice received with ZnNPs in combination with MA. The mRNA expression level of iNOS, TNF-α, and IFN-γ was dose-dependently (p < 0.01) upregulated; whereas it was downregulated in IL-10. ZnNPs markedly stimulated the caspase-3 activation with no significant toxicity on normal cells. Conclusion: Based on these in vitro and in vivo results, green synthesized ZnNPs, mainly along with MA, showed that has the potential to be introduced as a new drug for CL therapy. Triggering of NO production, and inhibition of infectivity rate are revealed as mechanisms of action ZnNPs on L. major. But, supplementary investigations are necessary to clear the efficacy and safety of these agents.
Article
Current treatment options for cutaneous leishmaniasis are associated with myriad limiting factors including low penetration, poor efficacy, and drug toxicities. Herein, we reported imiquimod and terbinafine co-loaded mannosylated transethosomes (IMQ-TER-MTES) with enhanced cutaneous retention, macrophage targeting, anti-leishmanial potential, and dermal immunomodulation. IMQ-TER-MTES were optimized using Design Expert® followed by their loading into chitosan gel. Moreover, the antileishmanial response against amastigotes-infected macrophages and Leishmania-infected BALB/c mice was evaluated. Finally, the safety and immunomodulation activity of IMQ-TER-MTES gel was performed using BALB/c mice. Optimized IMQ-TER-MTES showed nano-sized particles with low poly-dispersibility index (PDI) and high drug entrapment. Mannosylation has augmented macrophage targeting and the internalization capability of TES. IMQ-TER-MTES showed significantly reduced IC50 value (19.56 ± 3.62 μg/ml), higher selectivity index (29.24), and synergism against Leishmania major (L. major) amastigotes. In L. major infected BALB/c mice, the cutaneous lesion healing potential of IMQ-TER-MTES was also elevated with reduced lesion size (1.52 ± 0.43 mm). Superior safety of IMQ-TER-MTES was observed in BALB/c mice along with adequate stimulation of dermal immune cells, in contrast to the ALDARA®. Moreover, incremented Nuclear factor Kappa-β (NF-κβ) and nitric oxide (NO) biosynthesis were observed with IMQ-TER-MTES.
Article
Full-text available
Using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide microassay, previously described as a means of quantifying Leishmania amazonensis in vitro at the amastigote stage (D. Sereno and J. L. Lemesre, Parisitol. Res., in press), we have compared the activities of seven drugs, including those currently used to treat leishmaniasis, against axenically grown amastigote and promastigote forms of three Leishmania species (L. amazonensis, L. mexicana, and L. infantum, responsible for diffuse cutaneous, cutaneous, and visceral leishmaniasis, respectively). The ability of axenically cultured amastigote organisms to be used in an investigation of antileishmanial agents was first evaluated. We have confirmed the toxicities of sodium stibogluconate (Pentostam), pentamidine, and amphotericin B to active and dividing populations of axenically cultured amastigotes. The toxicity of potassium antimonyl tartrate trihydrate, which is generally higher than that of Pentostam, seemed to indicate that pentavalent antimony can be metabolized in vivo to compounds, possibly trivalent in nature, which are more active against the amastigote organisms. When the drug susceptibilities of parasites at both stages were compared, great variations were found for all the drugs studied. These major differences, which show the specific chemosusceptibility of the parasite at the mammalian stage, demonstrate the potential of using cultured amastigotes instead of promastigotes in a drug-screening procedure for early detection. This in vitro model may help in the isolation of active compounds, particularly those with low-grade activities, against the mammalian stage of the parasite.
Article
Full-text available
Inhibition of the major cytosolic protease, proteasome, has been reported to induce programmed cell death in several cell lines, while with other lines, similar inhibition blocked apoptosis triggered by a variety of harmful treatments. To elucidate the mechanism of pro- and antiapoptotic action of proteasome inhibitors, their effects on U937 lymphoid and 293 kidney human tumor cells were tested. Treatment with peptidyl aldehyde MG132 and other proteasome inhibitors led to a steady increase in activity of c-Jun N-terminal kinase, JNK1, which is known to initiate the apoptotic program in response to certain stresses. Dose dependence of MG132-induced JNK activation was parallel with that of apoptosis. Furthermore, inhibition of the JNK signaling pathway strongly suppressed MG132-induced apoptosis. These data indicate that JNK is critical for the cell death caused by proteasome inhibitors. An antiapoptotic action of proteasome inhibitors could be revealed by a short incubation of cells with MG132 followed by its withdrawal. Under these conditions, the major heat shock protein Hsp72 accumulated in cells and caused suppression of JNK activation in response to certain stresses. Accordingly, pretreatment with MG132 reduced JNK-dependent apoptosis caused by heat shock or ethanol, but it was unable to block JNK-independent apoptosis induced by TNFalpha. Therefore, proteasome inhibitors activate JNK, which initiates an apoptotic program, and simultaneously they induce Hsp72, which suppresses JNK-dependent apoptosis. A balance between these two effects might define the fate of cells exposed to the inhibitors.
Article
Full-text available
Background: Plant-derived flavonoids, which occur abundantly in our daily dietary intake, possess antitumor, antibacterial, and free radical scavenging properties. They form active constituents of a number of herbal and traditional medicines. Several flavonoids have been shown to exert their action by interacting with DNA topoisomerases and promoting site-specific DNA cleavage. Therefore, flavonoids are potential candidates in drug design. We report here that, although the flavonoids luteolin and quercetin are potent antileishmanial agents, luteolin has great promise for acting as a lead compound in the chemotherapy of leishmaniasis, a major concern in developing countries. Materials and methods: Kinetoplast DNA (kDNA) minicircle cleavage in drug-treated parasites was measured by electrophoresis of the total cellular DNA, followed by Southern hybridization using 32P labeled kDNA as a probe. Cell cycle progression and apoptosis were measured by flow cytometry using propidium iodide and fluorescein isothiocyanate (FITC)-labeled Annexin V. Results: Luteolin and quercetin inhibited the growth of Leishmania donovani promastigotes and amastigotes in vitro, inhibited DNA synthesis in promastigotes, and promoted topoisomerase-II-mediated linearization of kDNA minicircles. The IC50 values of luteolin and quercetin were 12.5 microM and 45.5 microM, respectively. These compounds arrest cell cycle progression in L. donovani promastigotes, leading to apoptosis. Luteolin has no effect on normal human T-cell blasts. Both luteolin and quercetin reduced splenic parasite burden in animal models. Conclusion: Luteolin and quercetin are effective antileishmanial agents. Quercetin has nonspecific effects on normal human T cells, but luteolin appears nontoxic. So, luteolin can be a strong candidate for antileishmanial drug design.
Article
Full-text available
Plant-derived flavonoids, which occur abundantly in the human diet, possess antitumour, antibacterial and free radical scavenging properties and form active constituents of a variety of herbal and traditional medicines. Several flavonoids have been shown to exert their action by interacting with DNA topoisomerases and promoting site-specific DNA cleavage. The flavonoids luteolin (obtained from Vitex negundo) and quercetin (obtained from Fagopyrum esculentum) are potent antileishmanial agents. Kinetoplast DNA (kDNA) minicircle cleavage in drug-treated Leishmania donovani was measured by electrophoresis of the total cellular DNA, followed by Southern hybridization using 32P labelled kDNA as a probe. Cell cycle progression and apoptosis were measured by flow cytometry using propidium iodide and fluorescein isothiocyanate (FITC)-labelled Annexin V. Luteolin and quercetin inhibited the growth of L. donovani promastigotes and amastigotes in vitro, inhibited DNA synthesis in promastigotes and promoted topoisomerase-II-mediated linearization of kDNA minicircles. The IC50 values of luteolin and quercetin were 12.5 and 45.5 µM, respectively. These compounds arrest cell cycle progression in L. donovani promastigotes, leading to apoptosis. Both luteolin (3.5 mg/kg twice weekly) and quercetin (14 mg/kg twice weekly) reduced splenic parasite burden in hamster models. Luteolin and quercetin are both effective antileishmanial agents. However, quercetin has nonspecific effects on normal human T cells although luteolin appears nontoxic. Luteolin appears to be a good candidate for antileishmanial drug design.
Article
Reactive nitrogen intermediates were synthesized spontaneously in cultures of macrophages from Trypanosoma brucei brucei -infected mice by an inducible nitric oxide (NO) synthase. This was inhibited by the addition of nitro- l -arginine. In this paper, we report the kinetics of the fixation of macrophage-derived NO on bovine serum albumin by using an enzyme-linked immunosorbent assay. S nitrosylation was confirmed by the Saville reaction, using mercuric chloride. It is known that reactive oxygen intermediates (ROI) are also synthesized by stimulated macrophages. The fact that NO is able to bind cysteine only under aerobic conditions led us to investigate the role of macrophage-derived ROI in the formation of S-nitrosylated proteins by activated macrophages. The immunoenzymatic signal decreased by 66 and 30% when superoxide dismutase and catalase, respectively, were added to the culture medium of macrophages from infected mice. In addition, the decrease in S-nitrosylated albumin formation correlated with the protection of extracellular trypanosomes from the cytostatic and cytotoxic activity of NO. Melatonin, a hydroxyl radical scavenger resulting from the decomposition of peroxynitrous acid, had no effect. All these data support the concept that an interaction between NO and ROI promoted the production of S -nitroso-albumin by activated macrophages from infected mice.
Article
The host response to Leishmania infection is regulated by a specific pattern of local cytokine production. We investigated the effect of interleukin (IL)-10 and IL-4 on the leishmanicidal activity of human macrophages (Mϕ). As with L. major, intracellular killing of L. infantum by human Mϕ was obtained following ligation of surface CD23 or cell treatment with Interferon-γ (IFN-γ). This leishmanicidal activity required nitric oxide (NO) generation by activated Mϕ, and it was partially mimicked by cell treatment with chemical NO donors. Addition of recombinant human IL-10 or IL-4 to CD23 mAb or IFN-γ decreased L. infantum and L. major killing by infected Mϕ. IL-10 was more potent than IL-4 in inhibiting the leishmanicidal activity of human Mϕ. Inhibition of Leishmania killing by IL-4 and IL-10 correlated with decreased NO generation from Mϕ, and was reversed when exogenous NO was added to cell cultures. Therefore, IL-10 and IL-4 down-regulate leishmanicidal activity of human Mϕ, in part by inhibiting NO generation by these cells.
Article
We have cloned the C. elegans cell death gene ced-3 . A ced-3 transcript is most abundant during embryogenesis, the stage during which most programmed cell deaths occur. The predicted CED-3 protein shows similarity to human and murine interleukin-1β-converting enzyme and to the product of the mouse nedd-2 gene, which is expressed in the embryonic brain. The sequences of 12 ced-3 mutations as well as the sequences of ced-3 genes from two related nematode species identify sites of potential functional importance. We propose that the CED-3 protein acts as a cysteine protease in the initiation of programmed cell death in C. elegans and that cysteine proteases also function in programmed cell death in mammals.
Article
Using plasma from patients infected or previously infected with Leishmania donovanii, we isolated a L. donovanii cDNA clone with similarity to the proteasome a-type subunit from humans and other eukaryotes. The cDNA clone, designated LePa, was DNA sequenced and Northern blot analysis of L. donovanii poly(A+)mRNA indicated the isolation of a full length cDNA clone with a transcript size of 1.9 kb. The expressed recombinant LePa fusion protein induced proliferation of peripheral blood mononuclear cells in one out of seven patients who had suffered from visceral leishmaniasis. Plasma from 16 out of 25 patients with visceral leishmaniasis and four out of 18 patients with cutaneous leishmaniasis contained IgG antibodies which reacted with the purified LePa fusion protein as evaluated in an ELISA. The LePa DNA sequence was inserted into an eukaryotic expression vector and Balb/c mice were vaccinated. DNA vaccination of Balb/c mice with LePa generated an initial significant reduction in lesion size after challenge.
Article
Apoptosis and/or programmed cell death have been described in examples ranging from fungi to man as gene-regulated processes with roles in cell and tissue physiopathology. These processes require the operation of an intercellular communicating network able to deliver alternative signals for cells with different fates and is thus considered a prerogative of multicellular organisms. Promastigotes from Leishmania (Leishmania) amazonensis, when shifted from their optimal in vitro growth temperature (22°C) to the temperature of the mammalian host (37°C), die by a calcium-modulated mechanism. More parasites die in the presence of this ion than in its absence, as detected by a colorimetric assay based on the activity of mitochondrial and cytoplasmic dehydrogenases which measures cell death, independently of the process by which it occurs. A heat shock, unable to induce detectable parasite death (34°C for 1 h), is able to significantly raise the concentration of intracellular free calcium in these cells. Heat-shocked parasites present ultrastructural and molecular features characteristic of cells dying by apoptosis. Morphological changes, observed only in the presence of calcium, are mainly nuclear. Cytoplasmic organelles are preserved. Heat shock is also able to induce DNA cleavage into an oligonucleosomal ladder detected in agarose gels by ethidium bromide staining and autoradiography of [32P]ddATP-labeled fragments. These results indicate that death by apoptosis is not exclusive of multicellular organisms. © 1996 Wiley-Liss, Inc.
Article
Nitric oxide (NO) releasing drugs (e.g., glyceryl trinitrate) were successfully used in the treatment of cutaneous leishmaniasis in man. In the present study, the effect of NO donors on the catalytic activity of the cysteine proteinase from promastigotes of Leishmania infantum, an agent of Old World visceral and cutaneous leishmaniases, is reported. In particular, one equivalent of NO, released by the NO donors S-nitrosoglutathione, glyceryl trinitrate, (±)-(E)-4-ethyl-2-[(E)-hydroxyimino]-5-nitro-3-hexenamide, 3-morpholinosydnonimine, S-nitrosoacetylpenicillamine and sodium nitroprusside, inhibited one equivalent of the parasite cysteine proteinase. As expected, NO-deprived compounds did not affect the catalytic activity of the parasite cysteine proteinase. Furthermore, the absorption spectrum of the (±)-(E)-4-ethyl-2-[(E)-hydroxyimino]-5-nitro-3-hexenamide-treated inactive L. infantum enzyme displayed a maximum in the 330–350 nm wavelength range. The reducing agents dithiothreitol and l-ascorbic acid completely prevented parasite cysteine proteinase inhibition by NO, fully restored the catalytic activity, and reversed the NO-induced absorption spectrum of the inactive enzyme. Moreover, S-nitrosoacetylpenicillamine displayed a leishmanicidal effect, inhibiting the cysteine proteinase activity in vivo. As expected, the NO-deprived compound N-acetylpenicillamine did not affect significantly the parasite viability and the enzyme activity in vivo. These data suggest that the L. infantum cysteine proteinase undergoes NO-mediated S-nitrosylation, thereby representing a possible mechanism of antiparasitic host defence.