Induces Long Lasting Intracellular Replication-Deficient: Leishmania Donovani

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Intracellular Replication-Deficient

Leishmania donovani Induces Long Lasting


Protective Immunity against Visceral
Leishmaniasis
This information is current as
of June 20, 2017. Angamuthu Selvapandiyan, Ranadhir Dey, Susanne Nylen,
Robert Duncan, David Sacks and Hira L. Nakhasi
J Immunol 2009; 183:1813-1820; Prepublished online 10
July 2009;
doi: 10.4049/jimmunol.0900276
http://www.jimmunol.org/content/183/3/1813

Downloaded from http://www.jimmunol.org/ by guest on June 20, 2017


Supplementary http://www.jimmunol.org/content/suppl/2009/07/13/jimmunol.090027
Material 6.DC1
References This article cites 42 articles, 26 of which you can access for free at:
http://www.jimmunol.org/content/183/3/1813.full#ref-list-1
Subscription Information about subscribing to The Journal of Immunology is online at:
http://jimmunol.org/subscription
Permissions Submit copyright permission requests at:
http://www.aai.org/About/Publications/JI/copyright.html
Email Alerts Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by


The American Association of Immunologists, Inc.,
1451 Rockville Pike, Suite 650, Rockville, MD 20852
Copyright © 2009 by The American Association of
Immunologists, Inc. All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
The Journal of Immunology

Intracellular Replication-Deficient Leishmania donovani


Induces Long Lasting Protective Immunity against Visceral
Leishmaniasis

Angamuthu Selvapandiyan,* Ranadhir Dey,* Susanne Nylen,† Robert Duncan,* David Sacks,†
and Hira L. Nakhasi1*
No vaccine is currently available for visceral leishmaniasis (VL) caused by Leishmania donovani. This study addresses whether a
live attenuated centrin gene-deleted L. donovani (LdCen1ⴚ/ⴚ) parasite can persist and be both safe and protective in animals.
LdCen1ⴚ/ⴚ has a defect in amastigote replication both in vitro and ex vivo in human macrophages. Safety was shown by the lack
of parasites in spleen and liver in susceptible BALB/c mice, immune compromised SCID mice, and human VL model hamsters 10
wk after infection. Mice immunized with LdCen1ⴚ/ⴚ showed early clearance of virulent parasite challenge not seen in mice
immunized with heat killed parasites. Upon virulent challenge, the immunized mice displayed in the CD4ⴙ T cell population a

Downloaded from http://www.jimmunol.org/ by guest on June 20, 2017


significant increase of single and multiple cytokine (IFN-␥, IL-2, and TNF) producing cells and IFN-␥/IL10 ratio. Immunized mice
also showed increased IgG2a immunoglobulins and NO production in macrophages. These features indicated a protective Th1-
type immune response. The Th1 response correlated with a significantly reduced parasite burden in the spleen and no parasites
in the liver compared with naive mice 10 wk post challenge. Protection was observed, when challenged even after 16 wk post
immunization, signifying a sustained immunity. Protection by immunization with attenuated parasites was also seen in hamsters.
Immunization with LdCen1ⴚ/ⴚ also cross-protected mice against infection with L. braziliensis that causes mucocutaneous leish-
maniasis. Results indicate that LdCen1ⴚ/ⴚ can be a safe and effective vaccine candidate against VL as well as mucocutaneous
leishmaniasis causing parasites. The Journal of Immunology, 2009, 183: 1813–1820.

T he obligate kinetoplastid protozoan parasites of the genus So far, several procedures have been used to develop live at-
Leishmania are spread by sand fly vectors and cause a tenuated Leishmania parasites including long-term in vitro cul-
spectrum of diseases collectively known as leishmaniasis tures, selection for temperature sensitivity, chemical mutagenesis,
that ranges from self-healing cutaneous leishmaniasis (CL),2 mu- and irradiation (1, 2). Although such live attenuated lines have
cocutaneous leishmaniasis (MCL), and to fatal visceral leishman- shown substantial protection against challenge in animal models,
iasis (VL). Leishmaniasis is endemic to 88 countries in the tropical undefined random genetic mutations and concerns arising from
and subtropical world, affecting 12 million people and threatening potential reversion to virulence make such vaccines unsuitable for
350 million more. Drug treatment requires long-term medication, human vaccination. Indeed, the persistence of asymptomatic in-
which is expensive and highly toxic. A vaccine for leishmaniasis fection especially in immunocompromised individuals raises the
has been a goal for a century, but there are still no effective vac- risk of reversion to clinical disease. Moreover, attenuations due to
cines (1). The knowledge that a cured disease either due to a nat- undefined genome alterations can reduce effective protective im-
ural infection or cutaneous leishmanization (1) protects the indi- munity, either they fail to persist long enough to elicit an immune
vidual from reinfection and the persistence of a few parasites in the response or lack critical epitopes to evoke the protective response
body can impart life-long protection against leishmaniasis, has en- (3). Alternatively, attenuation obtained through targeted genetic
couraged researchers to develop live attenuated Leishmania vac- disruptions of essential growth regulating or virulence genes by
cine candidates. homologous recombination is nonrevertible, hence can be safe (1).
Moreover, sustained exposure of parasite Ag to the host eliminates
the need of any adjuvants that are required in other nonliving vac-
*Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics cines (e.g., subunit vaccines).
Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, There are several examples of targeted gene deletions that
20892; and †Laboratory of Parasitic Diseases, National Institute of Allergy and In-
fectious Diseases, National Institutes of Health, Bethesda, MD 20892 have been conducted for developing Leishmania-attenuated
Received for publication January 27, 2009. Accepted for publication June 2, 2009.
vaccine strains. Among the vaccine candidates studied for CL,
L. major dihydrofolate reductase thymidylate synthase (dhfr-
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance ts⫺) knockout parasites protect mice (4) but not Rhesus mon-
with 18 U.S.C. Section 1734 solely to indicate this fact. keys (5). L. major deficient in surface and secreted phospho-
1
Address correspondence and reprint requests to Dr. Hira Nakhasi, Division of glycans (lpg2⫺), although unable to survive in sand flies and
Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and
Research, Food and Drug Administration, Bethesda, MD 20892. E-mail address:
macrophages, retained the ability to persist indefinitely in mice
hira.nakhasi@fda.hhs.gov and conferred protection against virulent challenge, even in the
2
Abbreviations used in this paper: CL, cutaneous leishmaniasis; MCL, mucocutane- absence of a strong Th1 response (6, 7). Over time, however,
2 ⫺
ous leishmaniasis; VL, visceral leishmaniasis; Wt, wild type; FTAg, freeze-thaw Ag; lpg (a complete gene knockout) parasites unexpectedly re-
WI, week immunized; WPC, week post challenge.
gained virulence (8). L. major deleted for phosphomanno-
Copyright © 2009 by The American Association of Immunologists, Inc. 0022-1767/09/$2.00 mutase-protected mice, despite no increase in either effector or

www.jimmunol.org/cgi/doi/10.4049/jimmunol.0900276
1814 REPLICATION DEFICIENT Leishmania INDUCES PROTECTIVE IMMUNITY

memory response (9). In contrast, L. mexicana that also causes an additional confirmation of the presence of parasites in tissues, total DNA
CL, deficient in cysteine proteinase genes (⌬cpa and ⌬cpb), samples obtained from infected mouse and hamster spleens were used as
conferred protection in mice and hamsters against homologous templates in a Taqman-based real-time PCR. The amplification target was
on the kinetoplast minicircle DNA of the parasite. The primers and meth-
challenge (10, 11). Among the vaccination studies in VL, mice ods were as previously described (22), with the addition of a fluorescent
immunized with a L. donovani strain deleted for biopterin trans- probe for detection. The probe had the sequence 5⬘-RAAARKKVRTRCA
porter (BT1) were similarly protected (12). Recent attempts us- GAAAYCCCGT-3⬘. A Black Hole Quencher moiety is coupled to the 3⬘
ing partial knockout parasites A2-A2rel gene cluster in L. do- end and Calfluor Red is coupled to a C6 linker at the 5⬘ end. The degenerate
letter code is according to the Nomenclature Committee of the Interna-
novani (13) and SIR2 gene in L. infantum (14) as immunogens tional Union of Biochemistry (http://www.chem.qmul.ac.uk/iubmb/misc/
induced protection against virulent challenge in BALB/c mice. naseq.html). The degenerate probe allows detection of sequence variants of
However, such mutants cannot be used as vaccine candidates, the minicircle found in Leishmania and is added to the reaction mixture at
because they still carry wild-type (Wt) allele/s and could cause a final concentration of 1.5 pmols/␮l. To evaluate the number of Leish-
disease. mania cells that were represented by a given Ct value, a standard curve was
constructed by infecting macrophages ex vivo (macrophage CSF cytokine
Despite limitations with most of the mutant strains, these ex- differentiated primary human monocytes) with stationary phase Leishma-
periments clearly demonstrate the potential as well as the pitfalls nia. After 24 h, macrophages were trypsinized, scraped into PBS, the mac-
of generating live attenuated Leishmania vaccines by targeted gene rophage cells/␮l were determined by hemocytometer, and the Leishmania
deletions. Hence it is critical to develop attenuated lines through amastigotes per macrophage were determined microscopically from a
smear of Diff-Quick (Baxter Healthcare Corporation) stained cells. A cal-
complete gene knockouts that generate avirulent organisms that culated volume of the cell suspension was added to DirectPCR Lysis Re-
persist for a brief duration but eventually are completely elimi- agent (Viagen Biotech) to produce lysates of 10 parasites/␮l, 1 parasite/␮l,
nated, thereby inducing effective immunity without clinical disease and 0.1 parasite/␮l. One microliter of each concentration was used as tem-
or the risk of reactivation. Recently, we have developed a L. do- plate in real-time minicircle PCR with four replicates to determine the

Downloaded from http://www.jimmunol.org/ by guest on June 20, 2017


novani strain completely deleted for the centrin1 gene (15). Cen- mean Ct value.
trin1 is a calcium-binding basal body-associated protein involved Intracellular staining and flow cytometry
in cell division in protozoan parasites like Leishmania, Trypano- Splenocytes were plated in 24-well plates and stimulated with freeze-thaw
soma, and Plasmodium (16 –18) and centrosome associated in Ag (FTAg) (23) or no Ag (control) in complete RPMI 1640 medium. After
higher eukaryotes (19). We demonstrated that deletion of LdCen1 36 h at 37°C brefeldin A was added to the wells. After 7 h at 37°C, cells
(LdCen1⫺/⫺) did not affect the growth of the promastigote in vitro, were blocked with anti-CD 16/32 (5 ␮g/ml) for 20 min (4°C), surface
however, the growth of the mammalian-infecting, amastigote form stained with PerCP anti-CD3, allophycocyanin-Cy7 anti-CD4, and PE-Cy7
anti CD8 Abs for 30 min (each with 1/300 dilution; 4°C), fixed with Cyto-
of the parasite, was blocked. These amastigotes showed failure of fix/Cytoperm kit for 20 min (room temperature), intracellular staining with
basal body duplication and cytokinesis, resulting in large multinu- Alex Flour 700 anti-IFN-␥, FITC anti-TNF, Pacific blue anti-IL-2, and
cleated cells in culture and ex vivo in human macrophages (15). APC anti-IL-10 or isotype-specific control for each fluorescence tagged Ab
We tested this unique parasite line for its safety and protective was for 30 min (each with 1/300 dilution; 4°C). Isotype controls for each
Ab used under similar conditions indicated specific binding of the test Ab.
efficacy against homologous and heterologous virulent Leishmania All Abs and reagents were purchased from BD Biosciences except Pacific
challenge and the immune response correlating with protection in blue anti-IL-2, which was from eBioscience. Electronic compensation was
rodent animal models. performed with single-stained cells with individual mAbs used in the test
samples. Cells were acquired on LSRII with DIVA software (BD Bio-
Materials and Methods sciences) and analyzed by FlowJo software (Tree Star).
Animals and parasites Ab responses
Five- to six-wk-old female BALB/c mice from National Cancer Institute, Specific Ab responses were measured by conventional ELISA. In brief,
SCID mice (BALB/c background) from The Jackson Laboratory, and ELISA plates were coated overnight at room temperature with FTAg. A
40 –50 gm male Syrian golden hamsters from Charles River Laboratories serial dilution of the sera was conducted to determine the titer, which is
were used in the experiments. Procedures used were reviewed and ap- defined as the inverse of the highest serum dilution factor giving an ab-
proved by the Animal Care and Use Committee, Center for Biologics Eval- sorbance of ⬎0.2. The titers for the Abs were determined using the fol-
uation and Research, Food and Drug Administration. Among parasites, the lowing HRP-conjugated secondary Abs: Rabbit anti-mouse IgG (H⫹L)-
Wt, centrin1-deleted (LdCen1⫺/⫺) and centrin1 deleted but episomally HRP; Rabbit anti-mouse IgG1-HRP, Human Adsorbed, Rabbit anti-mouse
centrin1 protein expressing line (LdCen1⫺/⫺ AB) of L. donovani (15) were IgG2a-HRP; Human Adsorbed (Southern Biotechnology Associates; all
used. The parasite culture procedure and the routine molecular biology with 1/1000 dilutions). SureBlue (KPL) was used as a peroxidase substrate.
practices were as previously described (16). After 15 min, the reaction was stopped by the addition of 100 ␮l of 1M
H2SO4, and the absorbance was read at 450 nm.
Vaccinations and challenge studies
NO quantifications
In independent experiments, the mice were inoculated/vaccinated via tail
vein with 3 million metacyclic cells of either L. donovani Wt, or Ld- Splenocytes or macrophages obtained from peritoneal fluid (24) were cul-
Cen1⫺/⫺ parasites. Infective-stage metacyclic promastigotes of L. dono- tured in complete, endotoxin-free RPMI 1640 medium (certified LPS free
vani were isolated from stationary cultures by density gradient centrifuga- by the manufacturer). NO (nitrite/nitrate) production was determined for
tion as described (20). Control groups (naive) corresponded to mice that the supernatants from unstimulated cultures and cultures stimulated with
received a saline solution (PBS). Mice vaccinated with LdCen1⫺/⫺ para- FTAg for 48 h at 37°C by the Griess reaction kit (Sigma-Aldrich).
sites after different time periods were challenged via tail vein with 3 million
virulent Wt L. donovani metacyclic parasites. Age-matched naive mice as Statistical analysis
control groups were also similarly challenged with 3 million virulent me- Statistical analysis of differences between means of groups was determined
tacyclic L. donovani parasites. In separate experiments, mice vaccinated by two-sample t test assuming unequal variance. A p value ⬍ 0.05 was
(with LdCen1⫺/⫺) or not (naive with saline) were also challenged by in- considered as highly significant.
jecting i.m. on left hind footpad with 3 million metacyclic L. braziliensis
parasites. Hamsters were inoculated intracardially with 10 million meta-
cyclics of Wt or LdCen1⫺/⫺ parasites. The hamsters vaccinated with Ld-
Results
Cen1⫺/⫺ or not (naive with saline) were challenged after 5 wk intracardi- Growth attenuation and limited persistence of LdCen1⫺/⫺
ally with 10 million metacyclic virulent L. donovani parasites. After post parasites in mice
challenge periods, parasite load was recorded from spleens and livers from
the L. donovani-challenged mice and hamsters and footpads and lymph Centrin1-deleted L. donovani metacyclics when used to infect hu-
nodes from the L. braziliensis-challenged mice by culturing the separated man macrophages in vitro or injected in susceptible BALB/c mice,
host cell preparations by limiting dilutions as previously described (21). As displayed an attenuated growth (i.e., reduction in the number of
The Journal of Immunology 1815

DNA from spleens of mice infected either with Wt or LdCen1⫺/⫺


parasites (supplementary Table 1). Detection of the parasite mi-
nicircle DNA target indicated the presence of a substantial number
of parasites in both Wt-infected (6 wk) and LdCen1⫺/⫺-infected (5
wk) mice. However, the mice infected with LdCen1⫺/⫺ for 12 wk
showed a level equivalent to naive mice, which can be considered
as a background, signifying either no LdCen1⫺/⫺ parasites or sig-
nificantly reduced parasite burden in mice at this time point.
Hence, it appears that the parasites are present for at least 5 wk in
the viscera as opposed to disappearing within just a few days.
Similarly, we did not observe any LdCen1⫺/⫺ parasites at 12 wk
postinfection in SCID mice, indicating that their clearance is not
dependent on functional T and B cells (Fig. 1b).

Protective efficacy of LdCen1⫺/⫺ parasites against virulent


infection
The ability of LdCen1⫺/⫺ parasite to protect against Leishmania
infection was determined in 5-wk immunized mice (WI) followed
by challenge with virulent Wt L. donovani. Spleen and liver were
analyzed for parasite burden 4, 8, and 10 wk post challenge

Downloaded from http://www.jimmunol.org/ by guest on June 20, 2017


(WPC), measured by limiting dilution. The results showed that
the immunized mice had ⬃2-log-fold reduced ( p ⬍ 0.01) parasite
burden in spleen and undetectable parasite in liver compared with
naive-challenged mice at 10 WPC (Fig. 1c). However, there was
no significant change in the weight of liver or spleen of immunized
or unimmunized mice. To evaluate the ability of LdCen1⫺/⫺ im-
munization to confer lasting protection, mice were challenged with
virulent Wt L. donovani 12 or 16 wk following LdCen1⫺/⫺ im-
munization, and evaluated after 10 wk. The parasite burden in the
spleen and liver indicated that the level of protection in both im-
FIGURE 1. Avirulence and immunoprotective properties of centrin munized groups (12 WI and 16 WI) was similar to the level ob-
(LdCen1) knockout L. donovani (LdCen1⫺/⫺) parasites in mice. a, Sur- served for the 5 WI group, indicating a sustained protective re-
vival of parasites of LdCen1⫺/⫺, LdCen1⫺/⫺ episomally expressing sponse after immunization (Fig. 1c). A similar result was observed
LdCen1 protein (add back; LdCen1⫺/⫺ AB), and Wt in BALB/c mice. in a separate experiment where mice were challenged after 24 wk
Parasite burdens from the organs of mice infected with metacyclic parasites
of immunization (data not shown). No protection was observed 8
at 5 wk post infection are shown. b, Centrin knockout L. donovani does not
WPC in mice immunized with heat killed 3 million LdCen1⫺/⫺
survive for long in BALB/c and SCID (BALB/c) mice. Parasite load in
organs of mice infected with metacyclics of either Wt or LdCen1⫺/⫺ par- metacyclics challenged 5 wk after immunization (supplementary
asites were measured at 5 or 12 wk after infection. c, Protection of Ld- Fig. 1b), suggesting that live attenuated parasites were required to
Cen1⫺/⫺ vaccinated BALB/c mice against virulent challenge. Mice vac- induce protective immunity. Overall, the results suggest that mice
cinated with LdCen1⫺/⫺ parasites were challenged after different time immunized with LdCen1⫺/⫺ elicit a strong and sustained protec-
periods with virulent Wt L. donovani and the challenge parasite burdens tion against the virulent challenge.
from the organs were measured after different time points post challenge.
The data presented are representative of two or more experiments with Induction of multifunctional Th1 effector cells correlates with
similar results. Mean and SEM of four or more mice in each group are LdCen1⫺/⫺-induced protection
shown. Cha, challenged; Imm, immunized; ND, not detected; WI, wk after
The involvement of IFN-␥-producing Th1 cells in immunity
infection; WC, wk after challenge; ⴱ, p ⬍ 0.01.
against Leishmania is described (25). We also observed an ab-
solute requirement for IFN-␥ in LdCen1⫺/⫺-induced immunity.
IFN-␥ knockout mice immunized with LdCen1⫺/⫺ for 5 wk
infected macrophages over time; supplementary Fig. 1a),3 and the followed by challenge were not protected. Parasite burden from
number of parasites per spleen or liver 5 wk postinfection com- these mice was similar to naive-challenged control observed at
pared with control (Fig. 1a). This is the direct consequence of 10 wk post challenge (Fig. 2). To further investigate the cell-
centrin1 deficiency, because LdCen1⫺/⫺ cells expressing centrin1 mediated responses induced by LdCen1⫺/⫺ parasites and find a
protein from a transfected plasmid were rescued for growth both in correlate of protective immunity, we measured the Th1 cyto-
macrophages and in mice (supplementary Fig. 1a and Fig. 1a). To kines IFN-␥, TNF, and IL-2 produced by splenic CD4⫹ and
analyze the persistence of LdCen1⫺/⫺ in mice, BALB/c mice were CD8⫹ T cells from the BALB/c mice using multiparameter flow
injected with LdCen1⫺/⫺ or Wt parasites and were monitored up to cytometry. Spleen cells grown in vitro with or without freeze
12 wk post infection. Few LdCen1⫺/⫺ parasites were seen at 5 wk thaw Ags followed by multicolor staining were gated based on
post infection in spleen and liver, measured by limiting dilution, forward and side scatter in FlowJo to select only the lympho-
and by 12 wk, the parasites were completely cleared from both cyte population devoid of dead cells and other larger leukocytes
organs (Fig. 1b). The Wt-infected mice had significant parasite (Fig. 3a, left) and further gating of the lymphocytes for the
burden in these organs at both the time points (Fig. 1b). Similar CD3⫹ (Fig. 3a, middle) followed by gating of CD3⫹ in to
results were obtained in a confirmatory real-time PCR study using CD4⫹ and CD8⫹ T cells (Fig. 3a, right). Seven distinct popu-
lations of cytokine-producing cells were defined from the CD4⫹
3
The online version of this article contains supplemental material. and CD8⫹ T cells based on different combinations of IFN- ␥,
1816 REPLICATION DEFICIENT Leishmania INDUCES PROTECTIVE IMMUNITY

Induction of humoral response in the immunized mice


We evaluated the humoral response in the immune-challenged
mice. Sera from BALB/c mice taken 10 wk post challenge after
5, 12, or 16 immunization weeks were measured for Leishma-
nia-specific IgG, IgG1, and IgG2a responses. Results indicated
a higher level of all three Ab populations in the immune-chal-
lenged groups compared with the naive challenged groups (Fig.
4a). Importantly, the immunization led to a selective increase in
Th1-driven IgG2a Ab levels during infection ( p ⬍ 0.01). The
results indicate a higher Leishmania specific humoral immune
response generated by immunization with LdCen1⫺/⫺ that cor-
relates with the increase in the Th1 response.
FIGURE 2. Lack of protection of LdCen1⫺/⫺ vaccinated IFN-␥ knock-
out mice against virulent challenge. IFN-␥ knockout mice vaccinated with Increased NO production in spleen cells in the
LdCen1⫺/⫺ parasites were challenged after 5 wk with virulent Wt L. do- immune-challenged mice
novani and the challenge parasite burdens from organs were measured after Because the production of NO by macrophages is a key factor in
10 wk post challenge. Mean and SEM of four mice in each group are killing Leishmania, we determined the level of NO produced by
shown.
in vitro Ag restimulated splenocytes that included macrophages
derived from the immune-challenged mice by ELISA. Interest-
ingly, significantly higher ( p ⬍ 0.01) levels of the reactive NO

Downloaded from http://www.jimmunol.org/ by guest on June 20, 2017


radical (nitrite) were demonstrated in the supernatants of
TNF, and IL-2 (a representative analysis is shown in Fig. 3b). splenocytes of the three immune-challenged groups (Fig. 4b).
The quality of the Th1 response is based on the relative fre- We also analyzed Ag-specific release of nitrite from the cul-
quency of these distinct populations (26). Five weeks after im- tured macrophages derived from the peritoneal fluids of mice.
munization with LdCen1⫺/⫺, single cytokine-producing CD4⫹ There is a small increase in NO production in macrophages of
and CD8⫹ T cells, making either IFN-␥ or IL-2, were more the naive mice, suggesting a background level in this assay, as
predominant than multicytokine-producing subpopulations post well as in the naive challenged mice with the addition of FTAg.
immunization (Fig. 3, c and d). Similar analysis conducted with However, in the immunized, challenged mice the production of
spleens of immunized mice analyzed 10 WPC showed a signif- NO in response to FTAg is significantly higher ( p ⬍ 0.01) than
icantly higher percent (2- to 10-fold; p ⬍ 0.01) of the cytokine- the naive challenged mice, which clearly correlates with pro-
producing subpopulations than in the naive challenged mice tection (supplemental Fig. 2).
(Fig. 3, e and f). Importantly, the expansion of multifunctional
cells was clearly evident in the immunized mice 10 WPC. In- Safety and efficacy of LdCen1⫺/⫺ in the hamster model
terestingly, the percent of both CD4⫹ and CD8⫹ T cells that Because golden Syrian hamsters are considered a more appro-
make TNF alone increased significantly (up to 2-fold; p ⬍ 0.01) priate model for VL (29), we evaluated the safety and protec-
after challenge in LdCen1⫺/⫺ immunized mice. The percentage tion of LdCen1⫺/⫺ against L. donovani challenge in this spe-
of T cells that produce Th1 cytokines increased after immuni- cies. The LdCen1⫺/⫺ parasite burden in spleens, measured by
zation and the increased frequency of multifunctional cells after limiting dilution, was 3-log-fold less than Wt L. donovani 5 wk
challenge strongly correlated with protection. after inoculation in hamsters and no LdCen1⫺/⫺ parasites were
We also quantitated T cells that produce IL-10, a cytokine in- found in the liver, whereas at 10 wk post immunization, no
volved in the pathogenesis of VL (27). The ratio of IFN-␥ to IL-10 LdCen1⫺/⫺ parasites were observed in any of the organs in
serves as an additional correlate of immune protection (14). In the contrast to hamsters inoculated with virulent parasites, which
restimulated CD4⫹ T cells from the spleen, the IFN-␥/IL-10 ratio had significantly higher number of parasites in both spleen and
was significantly higher in the immunized mice both at the time of liver (Fig. 5a). Further persistence of the parasites in the ham-
challenge (5 WI) and after challenge (5 WI plus 10 WC) compared ster model was evaluated by performing real-time minicircle
with either naive or naive-challenged controls (Fig. 3g). The re- PCR on spleen DNA samples collected more than 3 mo postin-
sults thus indicate an increased IFN-␥ secretion coinciding with fection. Samples from Wt-infected hamsters gave a Ct value
reduced IL-10 production among the immunized mice, indicating indicating a substantial number of parasites, while at the same
a strong Th1 response that could be accountable for protective time point, LdCen1⫺/⫺-infected samples gave a Ct value equiv-
immunity. alent to the uninfected hamsters. After 5 wk of immunization
Recent studies in CL infection in mice indicate a role for with LdCen1⫺/⫺ and challenge with virulent L. donovani, the
nonregulatory T cells, which simultaneously produce IFN-␥ and immunized hamsters had significantly lower parasite burden in
IL-10, for immune suppression during pathogenesis of a non- both the organs, measured by limiting dilution, compared with
healing lesion (28). Hence, we also looked at IFN-␥⫹ T cells the naive-challenged animals, with a reduction of 99.9% in
that coproduce IL-10 cytokine after immunization with Ld- spleen and 99.7% in liver ( p ⬍ 0.01) (Fig. 5b).
Cen1⫺/⫺. The results indicated a significantly higher percent-
age of CD4⫹ T cells positive for both IFN-␥ and IL-10 in the Centrin1 KO L. donovani cross protects mice against challenge
5-wk-immunized mice than in controls (Fig. 3h). No increase of with L. braziliensis
such cells in the CD4⫹ population was observed either 12 or 16 We wanted to determine whether immunization with Ld-
wk after immunization (data not shown). The presence of IFN-␥ Cen1⫺/⫺ could provide heterologous protection against infec-
and IL-10 coproducing CD4⫹ cells early after immunization tion involving other Leishmania species. For this purpose, 5 wk
(5W, a time point when LdCen1⫺/⫺ parasites are still detect- LdCen1⫺/⫺ immunized as well as naive BALB/c mice were
able), might reflect immune modulation to reduce inflamma- challenged by s.c. inoculation in the footpad with 2 million
tion-mediated host damage. metacyclics purified from L. braziliensis, the causative agent of
The Journal of Immunology 1817

Downloaded from http://www.jimmunol.org/ by guest on June 20, 2017


FIGURE 3. Multiparameter flow cytometry based analysis for distinct Th1/Th2 cytokine populations. A seven color cytometry panel was used to
measure the four cytokines, IFN-␥, TNF, IL2, and IL10, with the help of three T cell membrane markers CD3, CD4, and CD8. a, The common gating
steps shown in this study through FlowJo analysis are as explained in the Results section. b, Representative fluorescence intensity distributions of
single, double, or triple positive cytokines (IFN-␥, TNF, and IL-2) expressing T cells in spleens of vaccinated (5 wk post vaccination) and naive
mice. CD3⫹CD4⫹ or CD3⫹CD8⫹ lymphocytes from spleens showing either IFN-␥⫹ or IFN-␥⫺ are each further separated into cells that also make
either TNF, IL-2, both TNF and IL-2, or none. c–f, Flow cytometric analysis of Leishmania Ag-specific multifunctional T cells. Leishmania
Ag-specific multifunctional effector cells that were measured from splenocytes of LdCen1⫺/⫺ vaccinated mice at the time of challenge (5WI, 5 wk
post immunization) (c and d) and after challenge (5WI plus 10WPC: 10 wk post challenge) (e and f). The percentages of CD4⫹ (c and e) and CD8⫹
(d and f) T cells producing the cytokines IFN-␥, TNF, and IL-2 and combinations of these. The analysis was conducted using multiparameter flow
cytometry (as in Methods). g, Ratio of IFN-␥ to IL-10 producing CD4⫹ T cells from spleens at the time of challenge (naive and immunized (5W))
and after challenge (naive-challenged and immune-challenged (5WI plus 10WPC)). h, CD4⫹ T cells from spleens that simultaneously produce IFN-␥
and IL-10 at 5W post immunization with LdCen1⫺/⫺. The data presented are representative of two or more experiments with similar results.
Mean and SEM of four or more mice in each group with error bar indicating SEM are shown. Cha, challenged; Imm, immunized; W, wk infected;
WI, wk after immunization; WPC, wk after challenge; ⴱ, p ⬍ 0.01.

MCL. The naive, challenged mice developed progressive le- lenged mice showed significantly lower parasite burdens both in
sions during the 7 wk of observation, whereas the immunized the footpads and lymph nodes compared with the controls (Fig.
mice developed significantly smaller lesions (⬍3-fold; p ⬍ 5d), with a 99.9% reduction in both the tissues ( p ⬍ 0.02; p ⬍
0.01) throughout the 7 wk (Fig. 5c). The immune-chal- 0.01). A similar type of experiment conducted to examine cross
1818 REPLICATION DEFICIENT Leishmania INDUCES PROTECTIVE IMMUNITY

FIGURE 5. Avirulence and immuno-protective properties of


LdCen1⫺/⫺ parasites in hamsters. a, Parasite load in organs of hamsters
infected with either Wt or LdCen1⫺/⫺ metacyclic parasites was measured
at 5 and 10 wk after infection. b, Protection of the LdCen1⫺/⫺ vaccinated
hamsters against virulent challenge. Hamsters vaccinated with LdCen1⫺/⫺

Downloaded from http://www.jimmunol.org/ by guest on June 20, 2017


parasites were challenged after 5 wk of immunization with virulent Wt L.
donovani and the challenge parasite burdens from the organs were mea-
sured after 4 wk. c, Cross-protection of BALB/c mice vaccinated with
LdCen1⫺/⫺ against heterologous challenge with L. braziliensis. Five wk
post vaccinated mice were infected with metacyclics of L. braziliensis in-
jecting i.m. in the left-hind footpad of each mouse. Graph shows footpad
swelling due to infection in the injected footpad calculated by measuring
the difference in the footpad size between the two hind footpads. d, Parasite
numbers per organ of infected footpad or draining lymph nodes from 7 wk
postchallenge with L. braziliensis are shown. The data presented are rep-
resentative of two experiments with similar results. Mean and SEM of four
animals in each group are shown. Cha, challenged; Imm, immunized; ND,
not detected; W, wk after infection; ⴱ, p ⬍ 0.02 and ⴱⴱ, p ⬍ 0.01.
FIGURE 4. a, Humoral response in the virulent challenged mice vac-
cinated with LdCen1⫺/⫺ parasites. ELISA measurement of IgG, IgG1, and
IgG2a Abs in sera from the mice immunized with LdCen1⫺/⫺ parasites for To evaluate it as a vaccine candidate, the LdCen1⫺/⫺ parasite
5, 12, or 16 wk or naive before challenge with virulent L. donovani. Sera
was tested both in susceptible BALB/c mice and Syrian hamsters.
were collected 10 wk post challenge. b, Leishmania Ag specific stimulation
LdCen1⫺/⫺ parasites showed persistence of low numbers of par-
of NO synthase (NOS2) in the splenocytes of naive, naive challenged and
LdCen1⫺/⫺ immunized and Wt challenged mice. The activity of NOS2, asites both in mice and hamsters for at least 5 wk. Ten (both in
indicated by the amount of released nitrite (NO) in the splenocyte super- mice and hamsters) or 16 wk postinfection and in some cases even
natants was measured by the Griess reaction. The data presented are rep- longer periods (24 wk; data not shown), LdCen1⫺/⫺ parasites were
resentative of two experiments with similar results. Mean and SEM of three not detected in liver or spleen of mice, suggesting the complete
or more mice in each group are shown. Cha, challenged; Imm, immunized; clearance demanded of a safe vaccine candidate. Similar observa-
FTAg, Freeze-thaw Ag; WI, wk after immunization; WPC, wk after chal- tions in SCID mice lacking both T and B cells reinforce its safety
lenge; ⴱ, p ⬍ 0.01. even in the immune compromised host.
While searching for correlates of protection against homologous
challenge, we found that LdCen1⫺/⫺ immunized mice developed
an increased percent of Leishmania specific CD4⫹ and/or CD8⫹ T
protection of LdCen1⫺/⫺ against L. major showed a moderate cells expressing Th1 cytokines (IFN-␥, TNF, and IL-2) either sin-
but delayed protection (data not shown). gly or in multiple combinations. Multifunctional effector cells as-
sociated with protection have been described by others in mice
Discussion vaccinated with Leish-111f recombinant polyproteins plus adju-
In pursuit of a candidate live attenuated vaccine, we generated a L. vant (30) and with Leishmania MML protein expressed in a rep-
donovani parasite that had a complete deletion for a cell division lication-defective adenovirus (26). We found an especially strong
gene, centrin1 (LdCen1⫺/⫺) (15). We have demonstrated that increase of TNF-producing T cells, reinforcing prior data suggest-
LdCen1⫺/⫺ attenuation is due to the failure of cytokinesis, result- ing that TNF along with IFN-␥ secretion are important cytokines
ing in multiplication of cellular organelles, cell enlargement, and for protection in vivo (26). We also observed an increased IFN-
eventual programmed cell death. We think that the genetically tar- ␥/IL-10 ratio among the CD4⫹ T cells in the LdCen1⫺/⫺ vacci-
geted and defined attenuation reduces the risk of reversal to viru- nated mice both at the time of and after challenge, revealing an-
lence, a concern generally raised for attenuated organisms that are other correlate of protection. A similar polarization to an increased
created by random genomic mutations. LdCen1 deletion specifi- IFN-␥ to IL-10 ratio in the splenocyte supernatant measured by
cally attenuated the amastigote stage of the parasite that replicates ELISA after L. infantum challenge in mice immunized with
inside macrophages, and has no effect on the growth of the pro- SIR2⫹/⫺ L. infantum was observed (14). We also observed an
mastigote form. This could be advantageous to grow large quan- increase of CD4⫹ T cells that coproduce IFN-␥ and IL10 during
tities of parasites for vaccine trials. the initial period (5W) of infection with LdCen1⫺/⫺, suggesting
The Journal of Immunology 1819

possible immune modulation at this time as also observed previ- cies of Leishmania has been documented in animal model studies
ously, during L. major infection in mice (28). In the L. major case, (37–39). In mice, experimental vaccination using dp72 protein iso-
the nonregulatory T cells (CD4⫹CD25⫺Foxp3⫺) that were the lated from L. donovani cross-protected against L. major infection
source of IL-10 (most of them also produced IFN-␥) were immu- (40) and immunization with exogenous Ags (LmSEAgs) of L. ma-
nosuppressive during protozoan infection for the reduction of in- jor cross-protected against L. donovani (41). Our results show that
fection mediated damage to host cells. Hence, the LdCen1⫺/⫺ im- LdCen1⫺/⫺-immunized mice were cross-protected to a high de-
munization may be facilitating a desirable balance in the immune gree against a heterologous challenge with L. braziliensis, that
response allowing brief parasite persistence that facilitates protec- causes MCL, and to some degree against L. major that causes CL.
tion without causing host damage. Therefore, our results confirm that the live attenuated parasites also
In the present study, we observed a robust Th1-specific serum confer resistance to infection with other species.
Ab (IgG2a) response in the immune-challenged mice, further sup- In the literature, several routes of administration of Leishmania
porting the observation that LdCen1⫺/⫺ induces a generalized Th1 vaccine candidates have been studied with varied degree of pro-
type response. In addition, the increased release of NO observed in tection (reviewed in Ref. 42). In the present study, we have used
the splenocyte culture derived from the immune-challenged mice the i.v. route in mice and intracardial route in hamster, however we
coincided with the increase of Leishmania specific T cells produc- recognize that in general a recommended route of administration
ing IFN-␥, TNF, and IL-2 cytokines. NO production induced by of a vaccine in humans is either intradermal or i.m. Future studies
Th1 cytokines is a main leishmanicidal mechanism of murine mac- in our laboratory will focus on defining the ideal route of admin-
rophages (31, 32). Thus, in the LdCen1⫺/⫺-immunized mice, NO istration for optimal protection for LdCen1⫺/⫺ parasites.
production by macrophages that are stimulated by Th1 cytokine In summary, this study demonstrates that in mice and hamsters,
producing T cells correlates with the control of infection. the live attenuated LdCen1⫺/⫺ is highly immunogenic and confers

Downloaded from http://www.jimmunol.org/ by guest on June 20, 2017


Importantly, immunization with LdCen1⫺/⫺ protected both a significant degree of protection against L. donovani and also is
mice and hamsters against virulent homologous challenge, as in- capable of inducing cross protection against L. braziliensis. The
dicated by significantly reduced parasite burdens in the organs of vaccine-elicited parasite Ag-specific Th1 responses coinciding
the immune-challenged mice and hamsters compared with naive with a robust Ab response and NO production, all strongly corre-
controls. The protective response was sustained for up to 24 wk lated to a sustained protection. This report thus presents a well
after immunization. Complete elimination of wild-type parasites characterized amastigote specific, live attenuated L. donovani can-
from the livers of LdCen1⫺/⫺ immunized mice after virulent chal- didate vaccine that has been evaluated for its in vivo persistence,
lenge, is superior to the protection induced by L. donovani immunological response, and protective (against homologous and
BT1⫺/⫺, the only other complete gene knockout strain reported to heterologous challenges) efficacy.
target VL, which reduced parasite level in liver by 75% of the level
in naive challenged mice (12). The dhfr-ts⫺ auxotropic L. major Acknowledgments
line was safe in both mice and rhesus monkeys but protected only We acknowledge Kimberly Beacht for technical assistance and Sanjai Ku-
the mice (4, 5). Because the immune responses due to dhfr-ts⫺ mar and Alain Debrabant for valuable suggestions.
were not studied in those animals, it would be difficult to know if
there were weakness in the mouse immune response that could Disclosures
have predicted failure in the monkey. However, the quality of The authors have no financial conflict of interest.
LdCen1⫺/⫺-induced immune response in mice and protection in
mice and hamsters encourage us to proceed with immune studies References
in higher animal models. The level of protection achieved by 1. Selvapandiyan, A., R. Duncan, A. Debrabant, N. Lee, G. Sreenivas, P. Salotra,
LdCen1⫺/⫺ was similar to the protection due to L. infantum and H. L. Nakhasi. 2006. Genetically modified live attenuated parasites as vac-
cines for leishmaniasis. Indian J. Med. Res. 123: 455– 466.
SIR2⫹/⫺ in mice (14). Because L. infantum SIR2⫹/⫺ is a partial 2. Silvestre, R., A. Cordeiro-da-Silva, and A. Ouaissi. 2008. Live attenuated Leish-
knockout, it may be at a greater risk of reversion and therefore is mania vaccines: a potential strategic alternative. Arch. Immunol. Ther. Exp. 56:
unlikely to be pursued as a vaccine candidate. Lpg2⫺/⫺, the only 123–126.
3. Streit, J. A., T. J. Recker, F. G. Filho, S. M. Beverley, and M. E. Wilson. 2001.
complete gene knockout that has reverted to virulence in mice, is Protective immunity against the protozoan Leishmania chagasi is induced by
also known to establish prolonged persistence in the host (7), subclinical cutaneous infection with virulent but not avirulent organisms. J. Im-
which may have permitted the acquisition of compensating muta- munol. 166: 1921–1929.
4. Titus, R. G., F. J. Gueiros-Filho, L. A. de Freitas, and S. M. Beverley. 1995.
tions. Because LdCen1⫺/⫺ does not persists in mice and hamsters Development of a safe live Leishmania vaccine line by gene replacement. Proc.
beyond 2 mo, such a reversal to virulence is less likely. Natl. Acad. Sci. USA 92: 10267–10271.
5. Amaral, V. F., A. Teva, M. P. Oliveira-Neto, A. J. Silva, M. S. Pereira,
There are several possible explanations for the protection by E. Cupolillo, R. Porrozzi, S. G. Coutinho, C. Pirmez, S. M. Beverley, and
LdCen1⫺/⫺ parasite vaccination. First, there could be residual par- G. Grimaldi, Jr. 2002. Study of the safety, immunogenicity and efficacy of at-
asite burden (beyond the detection sensitivity of our methods, tenuated and killed Leishmania (Leishmania) major vaccines in a rhesus monkey
(Macaca mulatta) model of the human disease. Mem. Inst. Oswaldo Cruz 97:
which would allow continued presence of Leishmania-specific ef- 1041–1048.
fector cells and maintain anti-Leishmania immunity (21, 33). Sec- 6. Spath, G. F., L. F. Lye, H. Segawa, D. L. Sacks, S. J. Turco, and S. M. Beverley.
ond, vaccination with LdCen1⫺/⫺ parasites could lead to genera- 2003. Persistence without pathology in phosphoglycan-deficient Leishmania ma-
jor. Science 301: 1241–1243.
tion of a central memory T cell response after the KO parasites are 7. Uzonna, J. E., G. F. Spath, S. M. Beverley, and P. Scott. 2004. Vaccination with
cleared that could develop into an effector memory T cell response phosphoglycan-deficient Leishmania major protects highly susceptible mice from
upon challenge, and provide protection (34). Third, there could virulent challenge without inducing a strong Th1 response. J. Immunol. 172:
3793–3797.
be Ag persistence in peripheral tissues long after the parasites are 8. Spath, G. F., L. F. Lye, H. Segawa, S. J. Turco, and S. M. Beverley. 2004.
cleared. These persistent Ag “depots” may contribute to specific Identification of a compensatory mutant (lpg2-REV) of Leishmania major able to
survive as amastigotes within macrophages without LPG2-dependent glycocon-
memory T cells through the activation of naive T cells as has been jugates and its significance to virulence and immunization strategies. Infect. Im-
reported for some viral infections (35, 36). However, these hy- mun. 72: 3622–3627.
potheses need to be tested and are the subject of future studies. 9. Kedzierski, L., J. M. Curtis, P. C. Doherty, E. Handman, and K. Kedzierska.
2008. Decreased IL-10 and IL-13 production and increased CD44hi T cell re-
Finally, heterologous protection induced by infection with one cruitment contribute to Leishmania major immunity induced by non-persistent
virulent species of Leishmania against infection with another spe- parasites. Eur. J. Immunol. 38: 3090 –3100.
1820 REPLICATION DEFICIENT Leishmania INDUCES PROTECTIVE IMMUNITY

10. Alexander, J., G. H. Coombs, and J. C. Mottram. 1998. Leishmania mexicana Multifunctional TH1 cells define a correlate of vaccine-mediated protection
cysteine proteinase-deficient mutants have attenuated virulence for mice and po- against Leishmania major. Nat. Med. 13: 843– 850.
tentiate a Th1 response. J. Immunol. 161: 6794 – 6801. 27. Nylen, S., and D. Sacks. 2007. Interleukin-10 and the pathogenesis of human
11. Saravia, N. G., B. Escorcia, Y. Osorio, L. Valderrama, D. Brooks, L. Arteaga, visceral leishmaniasis. Trends Immunol. 28: 378 –384.
G. Coombs, J. Mottram, and B. L. Travi. 2006. Pathogenicity and protective 28. Anderson, C. F., M. Oukka, V. J. Kuchroo, and D. Sacks. 2007. CD4⫹CD25⫺
immunogenicity of cysteine proteinase-deficient mutants of Leishmania mexi- Foxp3⫺ Th1 cells are the source of IL-10-mediated immune suppression in
cana in non-murine models. Vaccine 24: 4247– 4259. chronic cutaneous leishmaniasis. J. Exp. Med. 204: 285–297.
12. Papadopoulou, B., G. Roy, M. Breton, C. Kundig, C. Dumas, I. Fillion, 29. Melby, P. C., B. Chandrasekar, W. Zhao, and J. E. Coe. 2001. The hamster as a
A. K. Singh, M. Olivier, and M. Ouellette. 2002. Reduced infectivity of a Leish- model of human visceral leishmaniasis: progressive disease and impaired gener-
mania donovani biopterin transporter genetic mutant and its use as an attenuated ation of nitric oxide in the face of a prominent Th1-like cytokine response. J. Im-
strain for vaccination. Infect. Immun. 70: 62– 68. munol. 166: 1912–1920.
13. Zhang, W. W., and G. Matlashewski. 2001. Characterization of the A2–A2rel
30. Coler, R. N., Y. Goto, L. Bogatzki, V. Raman, and S. G. Reed. 2007. Leish-111f,
gene cluster in Leishmania donovani: involvement of A2 in visceralization during
a recombinant polyprotein vaccine that protects against visceral Leishmaniasis by
infection. Mol. Microbiol. 39: 935–948.
elicitation of CD4⫹ T cells. Infect. Immun. 75: 4648 – 4654.
14. Silvestre, R., A. Cordeiro-Da-Silva, N. Santarem, B. Vergnes, D. Sereno, and
A. Ouaissi. 2007. SIR2-deficient Leishmania infantum induces a defined IFN-␥/ 31. Nathan, C. 1997. Inducible nitric oxide synthase: what difference does it make?
IL-10 pattern that correlates with protection. J. Immunol. 179: 3161–3170. J. Clin. Invest. 100: 2417–2423.
15. Selvapandiyan, A., A. Debrabant, R. Duncan, J. Muller, P. Salotra, G. Sreenivas, 32. Perez, L. E., B. Chandrasekar, O. A. Saldarriaga, W. Zhao, L. T. Arteaga,
J. L. Salisbury, and H. L. Nakhasi. 2004. Centrin gene disruption impairs stage- B. L. Travi, and P. C. Melby. 2006. Reduced nitric oxide synthase 2 (NOS2)
specific basal body duplication and cell cycle progression in Leishmania. J. Biol. promoter activity in the Syrian hamster renders the animal functionally deficient
Chem. 279: 25703–25710. in NOS2 activity and unable to control an intracellular pathogen. J. Immunol.
16. Selvapandiyan, A., R. Duncan, A. Debrabant, S. Bertholet, G. Sreenivas, 176: 5519 –5528.
N. S. Negi, P. Salotra, and H. L. Nakhasi. 2001. Expression of a mutant form of 33. Uzonna, J. E., G. Wei, D. Yurkowski, and P. Bretscher. 2001. Immune elimina-
Leishmania donovani centrin reduces the growth of the parasite. J. Biol. Chem. tion of Leishmania major in mice: implications for immune memory, vaccination,
276: 43253– 43261. and reactivation disease. J. Immunol. 167: 6967– 6974.
17. Selvapandiyan, A., P. Kumar, J. C. Morris, J. L. Salisbury, C. C. Wang, and 34. Zaph, C., J. Uzonna, S. M. Beverley, and P. Scott. 2004. Central memory T cells
H. L. Nakhasi. 2007. Centrin1 is required for organelle segregation and cytoki- mediate long-term immunity to Leishmania major in the absence of persistent

Downloaded from http://www.jimmunol.org/ by guest on June 20, 2017


nesis in Trypanosoma brucei. Mol. Biol. Cell 18: 3290 –3301. parasites. Nat. Med. 10: 1104 –1110.
18. Mahajan, B., A. Selvapandiyan, N. J. Gerald, V. Majam, H. Zheng, 35. Jelley-Gibbs, D. M., D. M. Brown, J. P. Dibble, L. Haynes, S. M. Eaton, and
T. Wickramarachchi, J. Tiwari, H. Fujioka, J. K. Moch, N. Kumar, et al. 2008. S. L. Swain. 2005. Unexpected prolonged presentation of influenza antigens pro-
Centrins, cell cycle regulation proteins in human malaria parasite Plasmodium motes CD4 T cell memory generation. J. Exp. Med. 202: 697–706.
falciparum. J. Biol. Chem. 283: 31871–31883. 36. Turner, D. L., L. S. Cauley, K. M. Khanna, and L. Lefrancois. 2007. Persistent
19. Salisbury, J. L. 1995. Centrin, centrosomes, and mitotic spindle poles. Curr. antigen presentation after acute vesicular stomatitis virus infection. J. Virol. 81:
Opin. Cell Biol. 7: 39 – 45. 2039 –2046.
20. Spath, G. F., and S. M. Beverley. 2001. A lipophosphoglycan-independent 37. Neal, R. A., A. Reeves, and W. Peters. 1990. Leishmania infecting man and wild
method for isolation of infective Leishmania metacyclic promastigotes by density animals in Saudi Arabia, 7: partial protection of mice against Leishmania major
gradient centrifugation. Exp. Parasitol. 99: 97–103. by prior infection with L. arabica. Trans. R. Soc. Trop. Med. Hyg. 84: 233–238.
21. Belkaid, Y., C. A. Piccirillo, S. Mendez, E. M. Shevach, and D. L. Sacks. 2002.
38. Gicheru, M. M., J. O. Olobo, and C. O. Anjili. 1997. Heterologous protection by
CD4⫹CD25⫹ regulatory T cells control Leishmania major persistence and im-
Leishmania donovani for Leishmania major infections in the vervet monkey
munity. Nature 420: 502–507.
model of the disease. Exp. Parasitol. 85: 109 –116.
22. Selvapandiyan, A., R. Duncan, J. Mendez, R. Kumar, P. Salotra, L. J. Cardo, and
H. L. Nakhasi. 2008. A Leishmania minicircle DNA footprint assay for sensitive 39. Lima, H. C., G. K. DeKrey, and R. G. Titus. 1999. Resolution of an infection with
detection and rapid speciation of clinical isolates. Transfusion 48: 1787–1798. Leishmania braziliensis confers complete protection to a subsequent challenge
23. Buxbaum, L. U., H. Denise, G. H. Coombs, J. Alexander, J. C. Mottram, and with Leishmania major in BALB/c mice. Mem. Inst. Oswaldo Cruz. 94: 71–76.
P. Scott. 2003. Cysteine protease B of Leishmania mexicana inhibits host Th1 40. Rachamim, N., and C. L. Jaffe. 1993. Pure protein from Leishmania donovani
responses and protective immunity. J. Immunol. 171: 3711–3717. protects mice against both cutaneous and visceral leishmaniasis. J. Immunol. 150:
24. Dey, R., A. Sarkar, N. Majumder, S. Bhattacharyya Majumdar, K. Roy- 2322–2331.
choudhury, S. Bhattacharyya, S. Roy, and S. Majumdar. 2005. Regulation of 41. Tonui, W. K., and R. G. Titus. 2007. Cross-protection against Leishmania do-
impaired protein kinase C signaling by chemokines in murine macrophages dur- novani but not L. braziliensis caused by vaccination with L. major soluble pro-
ing visceral leishmaniasis. Infect. Immun. 73: 8334 – 8344. mastigote exogenous antigens in BALB/c mice. Am. J. Trop. Med. Hyg. 76:
25. Scott, P. 1991. IFN-␥ modulates the early development of Th1 and Th2 responses 579 –584.
in a murine model of cutaneous leishmaniasis. J. Immunol. 147: 3149 –3155. 42. Bhowmick, S., T. Mazumdar, and N. Ali. 2009. Vaccination route that induces
26. Darrah, P. A., D. T. Patel, P. M. De Luca, R. W. Lindsay, D. F. Davey, transforming growth factor ␤ production fails to elicit protective immunity
B. J. Flynn, S. T. Hoff, P. Andersen, S. G. Reed, S. L. Morris, et al. 2007. against Leishmania donovani infection. Infect. Immun. 77: 1514 –1523.

You might also like