ArticlePDF Available

Chemokine Receptor CCR2 Is Not Essential for the Development of Experimental Cerebral Malaria

American Society for Microbiology
Infection and Immunity
Authors:

Abstract and Figures

Infection with Plasmodium berghei ANKA induces cerebral malaria in susceptible mice. Brain-sequestered CD8(+) T cells are responsible for this pathology. We have evaluated the role of CCR2, a chemokine receptor expressed on CD8(+) T cells. Infected CCR2-deficient mice were as susceptible to cerebral malaria as wild-type mice were, and CD8(+) T-cell migration to the brain was not abolished.
This content is subject to copyright. Terms and conditions apply.
INFECTION AND IMMUNITY, June 2003, p. 3648–3651 Vol. 71, No. 6
0019-9567/03/$08.000 DOI: 10.1128/IAI.71.6.3648–3651.2003
Copyright © 2003, American Society for Microbiology. All Rights Reserved.
Chemokine Receptor CCR2 Is Not Essential for the Development of
Experimental Cerebral Malaria
Elodie Belnoue,
1
Fabio T. M. Costa,
1
AnaM.Viga´rio,
1
Tatiana Voza,
2
Franc¸oise Gonnet,
1,2
Ire`ne Landau,
2
Nico van Rooijen,
3
Matthias Mack,
4
William A. Kuziel,
5
and Laurent Re´nia
1
*
De´partement d’Immunologie, Institut Cochin, INSERM U567, CNRS UMR 8104, Universite´ Rene´ Descartes, Hoˆpital Cochin,
1
and Museum National d’Histoire Naturelle,
2
Paris, France; Department of Cell Biology and Immunology, Faculty of
Medicine, Vrije Universiteit, Amsterdam 1081 BT, The Netherlands
3
; Medical Polyclinic, University of Munich,
80336 Munich, Germany
4
; and Department of Microbiology and Institute for Cellular and
Molecular Biology, University of Texas, Austin, Texas 78712-1095
5
Received 13 January 2003/Returned for modification 14 February 2003/Accepted 6 March 2003
Infection with Plasmodium berghei ANKA induces cerebral malaria in susceptible mice. Brain-sequestered
CD8
T cells are responsible for this pathology. We have evaluated the role of CCR2, a chemokine receptor
expressed on CD8
T cells. Infected CCR2-deficient mice were as susceptible to cerebral malaria as wild-type
mice were, and CD8
T-cell migration to the brain was not abolished.
Cerebral malaria (CM) contributes to around 2 million deaths
annually, mainly in African children. Brain sequestration of par-
asitized erythrocytes (PE) is thought to be responsible for this
pathology (4, 18). However, though necessary, PE sequestration
cannot account alone for CM, since this phenomenon has been
observed in non-CM cases (25). Leukocyte sequestration has of-
ten been described within brain postcapillary venules from pa-
tients who died of CM (9, 21); however, ethical considerations
limit investigation of the role of these cells in pathogenesis. In a
mouse model of CM with Plasmodium berghei ANKA, character-
ized by paralysis, deviation of the head, ataxia, convulsions, and
coma, histological studies have shown that PE and leukocytes are
sequestered in brain capillaries (10, 12, 20, 22). We have recently
demonstrated that recruitment of macrophages, neutrophils, and
T lymphocytes to the brain is associated with the onset of the
disease and that the recruited CD8
T-cell subset is responsible
for the neurological symptoms and the ensuing death (2). We
postulated that a chemokine receptor(s) must be necessary for
the migration of these pathogenic CD8
T cells to the brain. We
focused on one of these chemokine receptors, CCR2, since it has
been shown previously to be expressed on CD8
T cells migrating
to the brain after a viral infection (19). CCR2 is a member of the
seven-transmembrane G protein-coupled receptor superfamily
and binds ligands such as CCL2 (MCP-1), CCL7 (MCP-3), and
CCL12 (MCP-5) (29). In the mouse, CCR2 is expressed on
monocytes; T cells, in particular CD8
T cells (17); endothelial
cells; and brain cells like astrocytes and microglial cells (5, 11).
CCR2 has been shown elsewhere to be implicated in leukocyte
adhesion, monocyte recruitment (13, 26), and dendritic cell traf-
ficking (23).
With the use of a recently described monoclonal antibody
(MAb) to mouse CCR2 (17), expression of this molecule was
investigated by cytofluorometry on total brain-sequestered leuko-
cytes (BSL) and on the cell populations (macrophages and T
lymphocytes) which are known to express CCR2 (17), isolated
from 129/Ola C57BL/6J F
2
wild-type (WT) naive mice or P.
berghei ANKA-infected WT mice with or without CM. BSL were
isolated as previously described (2), and leukocyte subsets were
identified with the following antibodies: biotinylated rat immuno-
globulin G2b (IgG2b) MAb anti-mouse F4/80 (Tebu, Le Perray-
en-Yvelines, France), hamster IgG MAb anti-mouse CD3 conju-
gated to phycoerythrin (clone 17A2; PharMingen), rat IgG2a
antibody anti-mouse CD8conjugated to quantum red (clone
53-6.7; Sigma), rat IgG2a MAb anti-mouse CD4 conjugated to
quantum red (clone H129-19; Sigma), and purified rat antibody
anti-mouse CCR2 (17). Ultravidin conjugated to phycoerythrin
(Leinco Technologies Inc., St. Louis, Mo.) and goat IgG anti-rat
IgG conjugated to fluorescein isothiocyanate (Polysciences, Inc.,
Warrington, Pa.) were used as secondary reagents. For each sam-
ple, 5,000 cells were analyzed. CCR2
BSL were more numerous
in WT mice with CM than in those without CM (NCM) or in
naive mice (Fig. 1). BSL from WT mice with CM also expressed
more CCR2 on their surface (mean fluorescence intensity [MFI],
57.1 10.4) than did BSL from mice without CM (MFI, 30.1
2.9; one-factor analysis of variance and Tukey test, P0.05; five
mice per group) or BSL from naive mice (MFI, 25.15 2; P
0.01). Moreover, a strong and significant accumulation of CD8
T cells expressing CCR2 was observed in the brains of CM mice
but not in those from NCM or naive mice (Fig. 1B).
Since CCR2 is expressed on pathogenic CD8
T cells, we
next investigated susceptibility in CCR2-knockout (KO) mice
(14). These mice display severe deficits in macrophage (7, 14),
neutrophil (6), and T-cell (8) migration in response to either
antigenic or nonantigenic challenge and an impaired type 1
cytokine response (7). CCR2-KO and WT mice were infected
with 10
6
PE, and their parasitemia and anemia (hemoglobin
levels) were determined every other day as previously de-
scribed (28). All the KO mice but only 60 to 80% of WT mice
developed CM and died between days 6 and 10 after infection
(Fig. 2A and B). Though parasite levels were not significantly
different between the two groups during the first week, the
remaining WT mice died 2 weeks later (Fig. 2B) of hyperpara-
sitemia (Fig. 2C) and anemia (Fig. 2D).
* Corresponding author. Mailing address: De´partement d’Immuno-
logie, Institut Cochin, Hoˆpital Cochin, Baˆtiment Gustave Roussy, 27
rue du Fbg St Jacques, 75014 Paris, France. Phone: 33 1 40 51 65 11.
Fax: 33 1 40 51 65 35. E-mail: renia@cochin.inserm.fr.
3648
Histopathological analysis of the midbrain region of infected
mice was performed as described previously (1) and revealed
petechial hemorrhages and leukocyte accumulation in the cap-
illaries of WT mice with CM, whereas these changes were not
observed in infected WT mice without CM (data not shown).
Brains of infected KO mice with CM showed ring hemorrhages
with apparently fewer leukocytes in the capillaries than in
those of WT mice with CM (data not shown). We thus quan-
tied the total number of BSL from WT and KO mice. As
shown in Fig. 3, BSL from KO mice with CM were less nu-
merous than BSL from WT mice with CM. Nevertheless, there
was a signicant threefold increase in BSL number in infected
KO mice compared to naive KO mice. There were eight times
more BSL from WT mice with CM than from naive WT mice.
NCM WT mice contained the same number of BSL as did
naive WT mice (Fig. 3). BSL from the different mouse groups
were further phenotyped by cytouorometry. Macrophages
were identied as F4/80
, neutrophils were identied as F4/
80
and Gr-1
(rat IgG2b MAb anti-mouse Gr-1 conjugated
to uorescein isothiocyanate, clone RB6-8C5; PharMingen),
and T cells were identied as described above. We observed a
signicant increase in the numbers of macrophages, neutro-
phils, and CD4
and CD8
T lymphocytes (but not of other
cell types) in CM WT mice compared with naive or NCM WT
mice. Macrophages and CD8
T cells, but no other cell types,
increased in infected KO mice with CM compared with naive
KO mice. However, the number of macrophages in KO mice
with CM was signicantly lower than in CM WT mice (Fig. 3).
In contrast, similar numbers of CD8
T cells, the subset re-
sponsible for CM in WT mice, were found in CM WT and CM
KO mice. Depletion experiments were carried out to investi-
gate the role of brain-sequestered CD8
T cells in CCR2-KO
mice with CM. Depletion of BSL subsets was performed at day
6, just before the onset of CM, by injecting intraperitoneally 1
mg of the following MAbs: rat IgG anti-mouse CD8 (clone
2.43; ATCC TIB 210), rat IgG anti-mouse CD4 (clone GK1.5;
ATCC TIB 207), or antipolymorphonuclear cells (15). More
than 98% of blood CD8
or CD4
T cells were depleted as
veried by uorescence-activated cell sorting (FACS) analysis.
Depletion of blood neutrophils was more than 80% as veried
FIG. 1. CCR2 is expressed on BSL. (A) Representative dot plot of BSL from CCR2 WT mice (naive, NCM, and CM) stained with an
anti-CCR2 MAb versus size (forward size scatter [FSC]). Data are representative of ve animals per group. (B) Number of total sequestered
leukocyte subsets (white bars) and CCR2
leukocyte subsets (black bars) from the whole brain of WT mice (naive, NCM, and CM). Samples of
brain leukocyte suspension from mice infected with 10
6
PE and healthy mice were stained with MAbs specic for neutrophils, macrophages, and
CD4
and CD8
T cells and for CCR2 and analyzed by ow cytometry. Absolute numbers of a given subset were calculated by multiplying the
percentage of positive cells for this subset by the total number of BSL. CCR2
cell numbers were determined by using the percentage of CCR2
positive cells within each subset multiplied by the total number of this subset. *, P0.05 versus CM mice (one-factor analysis of variance followed
by Tukey test). #, P0.05 versus NCM mice. This experiment is representative of three.
VOL. 71, 2003 NOTES 3649
by FACS analysis with anti-Gr-1 MAb. Puried rat IgG (Sig-
ma) was used as a negative control. Macrophages were de-
pleted at day 5 after P.berghei ANKA injection by intravenous
injection of 0.2 ml of phosphate-buffered saline containing
approximately 1 mg of dichloromethylenediphosphonate (Cl
2
-
MDP) encapsulated in liposomes (27). More than 90% of
blood F4/80
cells were depleted as veried by FACS analysis
2 days later. All CCR2-KO mice depleted of CD4
T cells,
neutrophils, or macrophages died of CM (Fig. 4 and data not
shown), whereas none of the anti-CD8-treated KO mice de-
FIG. 2. CM incidence, survival, parasite load, and hemoglobin lev-
els after P.berghei ANKA (PbA) infection of CCR2 WT and KO mice.
(A) CM incidence occurring between day 6 and day 10 in WT (n32)
and KO (n27) mice infected with 10
6
PE. On day 10, as calculated
by Fishers exact test, Pwas 0.0049 between WT and KO mice. (B) Sur-
vival of WT (n17) and KO (n17) mice infected with 10
6
PE.
Neurological signs rst appear late on days 6 to 10 (shaded area), with
death occurring in 24 h after their onset. (C) PE per milliliter of
blood standard errors of the means. WT (n5) and KO (n5)
mice were infected with 10
6
PE. Mortality is indicated at the top (KO
mice) and at the bottom (WT mice) as the number of dead mice (d) on
that day. The difference between WT and KO mice on day 6 was not
signicant. (D) Hemoglobin levels (means standard errors of the
means) in WT (n5) and KO (n5) mice infected with 10
6
PE.
FIG. 3. Levels of whole-brain-sequestered leukocytes in CCR2-KO and WT mice after P.berghei ANKA infection. Enumeration of BSL was
performed on perfused brains from KO (n15) and WT (n8) mice at the time when CM is diagnosable (days 6 to 10), NCM WT mice (days
9 to 10) (n6), and naive KO (n10) or WT (n11) mice. Cell numbers were determined as described for Fig. 1B. Values are expressed as
means standard errors of the means. *, P0.05 (one-factor analysis of variance followed by Tukey test), signicantly different from naive WT
mice; #, P0.05, Tukey test, signicantly different from NCM WT mice; and , P0.05, Tukey test, signicantly different from CM KO mice.
This experiment is representative of three.
FIG. 4. Role of CD8
T cells in CM in CCR2 KO mice. The
effector role of CD8
T cells was demonstrated through a series of
depletion experiments with infected CCR2-KO and WT mice. The
gure shows survival (A) and CM incidence (B) in infected WT or KO
mice injected with the following rat antibodies: control IgG (n5),
anti-CD8 (n5), anti-CD4 (n5), and anti-polymorphonuclear cell
(PMN) (n5) on day 6. *, P0.05 (Fisher test) versus P.berghei
ANKA-infected WT mice treated with control rat IgG; #, P0.0001
(Fisher test) versus rat IgG-treated KO mice. This experiment is rep-
resentative of two.
3650 NOTES INFECT.IMMUN.
veloped CM. Identical results were observed in infected and
similarly depleted WT mice (Fig. 4 and data not shown).
Finally the role of cytokines was investigated, since a type 1
response, which is altered in CCR2-KO mice (7, 23, 24), has
been associated elsewhere with CM development (1, 16). Both
CM WT and KO mice, however, developed similar serological
and cellular type 1 responses overall (data not shown).
Our results clearly show that CCR2 is not necessary for CM to
occur. CCR2 deciency was associated with a reduction in num-
bers of macrophages, neutrophils, and CD4
T cells but not of
CD8
T cells. Our results further conrm that CD8
T cells are
responsible for CM death (2). It is remarkable that the pathology
in WT and CCR2-KO mice was due to the sequestration of less
than 10
5
CD8
T cells in the vasculature of a whole brain. CCR2
has also been shown previously to be expressed on brain cells like
endothelial cells, astrocytes, and microglial cells (5, 11), and sig-
naling through this receptor may activate these cell types for
chemokine and cytokine production. However, our results indi-
cate that CCR2 signaling in these cells is not required for the
development of CM. Since migration of CD8
T cells to the brain
occurred normally in CCR2 KO mice, this implies that another
chemokine receptor(s) is involved in this process. We have shown
recently that CCR5 deciency results in the decrease in CM
susceptibility in mice of the same genetic background (3). Prelim-
inary results indicate that more than 80% of brain-sequestered
CD8
T cells from infected WT or CCR2 KO mice express
CCR5 (data not shown). More studies are needed to determine if
other chemokine receptors are involved in rodent and eventually
in human CM.
We thank Georges Snounou for critical reading of the manuscript.
This work was supported in part by a grant from Junta Nacional de
Investigac¸a˜o Cientica e Tecnologica (JNICT) and Fondation de La
Recherche Me´dicale to Laurent Re´nia. Elodie Belnoue held a fellow-
ship from MENRT. Fabio T. M Costa was supported by a fellowship
from the CAPES foundation, Brazil. Ana Margarida Viga´rio held a
fellowship from Junta Nacional de Investigac¸a˜o Cientica e Tecno-
logica (JNICT), Portugal.
REFERENCES
1. Amani, V., A. M. Vigario, E. Belnoue, M. Marussig, L. Fonseca, D. Mazier,
and L. Re´nia. 2000. Involvement of IFN-receptor-mediated signaling in
pathology and anti-malarial immunity induced by Plasmodium berghei infec-
tion. Eur. J. Immunol. 30:16461655.
2. Belnoue, E., M. Kayibanda, A. M. Vigario, J. C. Deschemin, N. Van Rooijen,
M. Viguier, G. Snounou, and L. Renia. 2002. On the pathogenic role of
brain-sequestered ␣␤ CD8
T cells in experimental cerebral malaria. J. Im-
munol. 169:63696375.
3. Belnoue, E., M. Kayibanda, J.-C. Deschemin, M. Viguier, M. Mack, W. A.
Kuziel, and L. Renia. CCR5 deciency decreases susceptibility to experi-
mental cerebral malaria. Blood, in press.
4. Berendt, A. R., G. D. H. Turner, and C. I. Newbold. 1994. Cerebral malaria:
the sequestration hypothesis. Parasitol. Today 10:412414.
5. Berger, O., X. Gan, C. Gujuluva, A. R. Burns, G. Sulur, M. Stins, D. Way, M.
Witte, M. Weinand, J. Said, K. S. Kim, D. Taub, M. C. Graves, and M. Fiala.
1999. CXC and CC chemokine receptors on coronary and brain endothelia.
Mol. Med. 5:795805.
6. Blease, K., B. Mehrad, T. J. Standiford, N. W. Lukacs, J. Gosling, L. Boring,
I. F. Charo, S. L. Kunkel, and C. M. Hogaboam. 2000. Enhanced pulmonary
allergic responses to aspergillus in CCR2
/
mice. J. Immunol. 165:2603
2611.
7. Boring, L., J. Gosling, S. W. Chensue, S. L. Kunkel, R. V. Farese, Jr., and
H. E. Broxmeyer. 1997. Impaired monocyte migration and reduced type 1
(Th1) cytokine responses in C-C chemokine receptor 2 knockout mice.
J. Clin. Investig. 100:25522561.
8. Fife, B. T., G. B. Huffnagle, W. A. Kuziel, and W. J. Karpus. 2000. CC
chemokine receptor 2 is critical for induction of experimental autoimmune
encephalomyelitis. J. Exp. Med. 192:899905.
9. Grau, G. E., C. D. Mackenzie, R. A. Carr, M. Redard, G. Pizzolato, C.
Allasia, C. Cataldo, T. E. Taylor, and M. E. Molyneux. 2003. Platelet accu-
mulation in brain microvessels in fatal pediatric cerebral malaria. J. Infect.
Dis. 187:461466.
10. Hearn, J., N. Rayment, D. N. Landon, D. R. Katz, and J. B. De Souza. 2000.
Immunopathology of cerebral malaria: morphological evidence of parasite
sequestration in murine brain microvasculature. Infect. Immun. 68:5364
5376.
11. Hesselgesser, J., and R. Horuk. 1999. Chemokine and chemokine receptor
expression in the central nervous system. J. Neurovirol. 5:1326.
12. Jennings, V. M., J. K. Actor, A. A. Lal, and R. L. Hunter. 1997. Cytokine
prole suggesting that murine cerebral malaria is an encephalitis. Infect.
Immun. 65:48834887.
13. Kurihara, T., G. Warr, J. Loy, and R. Bravo. 1997. Defects in macrophage
recruitment and host defense in mice lacking the CCR2 chemokine receptor.
J. Exp. Med. 186:17571762.
14. Kuziel, W. A., S. J. Morgan, T. C. Dawson, S. Grifn, O. Smithies, K. Ley,
and N. Maeda. 1997. Severe reduction in leukocyte adhesion and monocyte
extravasation in mice decient in CC chemokine receptor 2. Proc. Natl.
Acad. Sci. USA 94:1205312058.
15. Lopez, A. F., M. Strath, and C. J. Sanderson. 1984. Differentiation antigens
on mouse eosinophils and neutrophils identied by monoclonal antibodies.
Br. J. Haematol. 57:489494.
16. Lou, J., R. Lucas, and G. E. Grau. 2001. Pathogenesis of cerebral malaria:
recent experimental data and possible applications for humans. Clin. Micro-
biol. Rev. 14:810820.
17. Mack, M., J. Cihak, C. Simonis, B. Luckow, A. E. Proudfoot, J. Plachy, H.
Bruhl, M. Frink, H. J. Anders, V. Vielhauer, J. Pstinger, M. Stangassinger,
and D. Schlondorff. 2000. Expression and characterization of the chemokine
receptors CCR2 and CCR5 in mice. J. Immunol. 166:46974704.
18. MacPherson, G. G., M. J. Warrell, N. J. White, S. Looareesuwan, and D. A.
Warrell. 1985. Human cerebral malaria. A quantitative ultrastructural anal-
ysis of parasitized erythrocyte sequestration. Am. J. Pathol. 119:385401.
19. Nansen, A., O. Marker, C. Bartholdy, and A. R. Thomsen. 2000. CCR2
and
CCR5
CD8
T cells increase during viral infection and migrate to sites of
infection. Eur. J. Immunol. 30:17971806.
20. Neill, A. L., and N. H. Hunt. 1992. Pathology of fatal and resolving Plasmo-
dium berghei cerebral malaria in mice. Parasitology 105:165175.
21. Patnaik, J. K., B. S. Das, S. K. Mishra, S. Mohanty, S. K. Satpathy, and D.
Mohanty. 1994. Vascular clogging, mononuclear cell margination, and en-
hanced vascular permeability in the pathogenesis of human cerebral malaria.
Am. J. Trop. Med. Hyg. 51:642647.
22. Rest, J. R. 1982. Cerebral malaria in inbred mice. I. A new model and its
pathology. Trans. R. Soc. Trop. Med. Hyg. 76:410415.
23. Sato, N., S. K. Ahuja, M. Quinones, V. Kostecki, R. L. Reddick, P. C. Melby,
W. A. Kuziel, and S. S. Ahuja. 2000. CC chemokine receptor (CCR)2 is
required for Langerhans cell migration and localization of Th1 cell type
(Th1)-inducing dendritic cells: absence of CCR2 shifts the Leishmania ma-
jor-resistant phenotype to a susceptible state dominated by Th2 cytokines, B
cell outgrowth, and sustained neutrophilic inammation. J. Exp. Med. 192:
205243.
24. Sato, N., W. A. Kuziel, P. C. Melby, R. L. Reddick, V. Kostecki, W. Zhao, N.
Maeda, S. K. Ahuja, and S. S. Ahuja. 1999. Defects in the generation of
IFN-are overcome to control infection with Leishmania donovani in CC
chemokine receptor (CCR) 5-, macrophage inammatory protein-1-, or
CCR2-decient mice. J. Immunol. 163:55195525.
25. Silamut, K., N. H. Phu, C. Whitty, G. D. Turner, K. Louwrier, N. T. Mai,
J. A. Simpson, T. T. Hien, and N. J. White. 1999. A quantitative analysis of
the microvascular sequestration of malaria parasites in the human brain.
Am. J. Pathol. 155:395410.
26. Traynor, T. R., W. A. Kuziel, G. B. Toews, and G. B. Huffnagle. 2000. CCR2
expression determines T1 versus T2 polarization during pulmonary Crypto-
coccus neoformans infection. J. Immunol. 164:20212027.
27. Van Rooijen, N., A. Sanders, and T. K. van den Berg. 1996. Apoptosis of
macrophages induced by liposome-mediated intracellular delivery of clod-
ronate and propamidine. J. Immunol. Methods 193:9399.
28. Vigario, A. M., E. Belnoue, A. Cumano, M. Marussig, F. Miltgen, I. Landau,
D. Mazier, I. Gresser, and L. Renia. 2001. Inhibition of Plasmodium yoelii
blood-stage malaria by interferon alpha through the inhibition of the pro-
duction of its target cell, the reticulocyte. Blood 97:39663971.
29. Zlotnik, A., and O. Yoshie. 2000. Chemokines: a new classication system
and their role in immunity. Immunity 12:121127.
Editor: W. A. Petri, Jr.
VOL. 71, 2003 NOTES 3651
... Many individual immune cell populations including neutrophils [27,28], macrophages/monocytes [18,29], NK cells [30], and CD4 + T cells [31,32] have been implicated in the pathogenesis of this disease. However, other studies have shown that neither antibody-mediated nor genetic depletion of these cells affected the accumulation of iRBCs in the CNS [33] or the ability of mice to develop ECM [21,[34][35][36]. Therefore, the contribution of these immune cell subsets to ECM is still a matter of debate. ...
... The contribution of innate immune cells to the pathogenesis of ECM is still a matter of contention. Previous studies have either implicated monocytes and neutrophils in the pathogenesis of ECM [18,[27][28][29] or shown them to be irrelevant [21,33,35,36]. Our flow cytometric ( Fig 1) and imaging ( Fig 2) data suggested that these cells might be involved in disease pathogenesis. ...
... This is different from the significant vascular disruption induced by synchronously extravasating myelomonocytic cells during fatal viral meningitis [42]. Some studies have implicated myelomonocytic cells in the pathogenesis of ECM [18,[27][28][29], whereas others have not [21,33,35,36]. The difference in the outcome of these studies could be linked to many different variables, including strain of mice, depletion strategy, inadvertent blockade of T cells, and genetic variation in the strain of Plasmodium used. ...
Article
Full-text available
Cerebral malaria (CM) is a severe complication of Plasmodium falciparum infection that results in thousands of deaths each year, mostly in African children. The in vivo mechanisms underlying this fatal condition are not entirely understood. Using the animal model of experimental cerebral malaria (ECM), we sought mechanistic insights into the pathogenesis of CM. Fatal disease was associated with alterations in tight junction proteins, vascular breakdown in the meninges / parenchyma, edema, and ultimately neuronal cell death in the brainstem, which is consistent with cerebral herniation as a cause of death. At the peak of ECM, we revealed using intravital two-photon microscopy that myelomonocytic cells and parasite-specific CD8⁺ T cells associated primarily with the luminal surface of CNS blood vessels. Myelomonocytic cells participated in the removal of parasitized red blood cells (pRBCs) from cerebral blood vessels, but were not required for the disease. Interestingly, the majority of disease-inducing parasite-specific CD8⁺ T cells interacted with the lumen of brain vascular endothelial cells (ECs), where they were observed surveying, dividing, and arresting in a cognate peptide-MHC I dependent manner. These activities were critically dependent on IFN-γ, which was responsible for activating cerebrovascular ECs to upregulate adhesion and antigen-presenting molecules. Importantly, parasite-specific CD8⁺ T cell interactions with cerebral vessels were impaired in chimeric mice rendered unable to present EC antigens on MHC I, and these mice were in turn resistant to fatal brainstem pathology. Moreover, anti-adhesion molecule (LFA-1 / VLA-4) therapy prevented fatal disease by rapidly displacing luminal CD8⁺ T cells from cerebrovascular ECs without affecting extravascular T cells. These in vivo data demonstrate that parasite-specific CD8⁺ T cell-induced fatal vascular breakdown and subsequent neuronal death during ECM is associated with luminal, antigen-dependent interactions with cerebrovasculature.
... Studies have shown that CCR2 is widely involved in conferring a protective immune response against the pathogen. CCR2 is a receptor that promotes the induction of several effector cells, such as activated "T cells," "NK cells," "dendritic cells," and "monocytes" to the site of infection [143].Nevertheless, it is also observed that genetic deficiency of CCR2 does not appear to affect parasite growth during P. berghei-ANKA [144] and Plasmodium chabaudi adami [145] infection, while CXCR3, CXCL9,and CXCL10 assist in the progress of cerebral malaria [88,89]. On the other hand, CCR2 -/mice infected with Plasmodium chabaudi chabaudi show a delay in clearance of the parasite [84]. ...
Article
Chemokines belong to the group of small proteins within the cytokine family having strong chemo-attractant properties. In most cases, the strong immuno-modulatory role of chemokines is crucial for generating the immune response against pathogens in various protozoan diseases. In this review, we have given a brief update on the classification, characterization, homeostasis, transcellular migration, and immuno-modulatory role of chemokines. Here we will evaluate the potential role of chemokines and their regulation in various protozoan diseases. There is a significant direct relationship between parasitic infection and the recruitment of effector cells of the immune response. Chemokines play an indispensable role in mediating several defense mechanisms against infection, such as leukocyte recruitment and the generation of innate and cell-mediated immunity that aids in controlling/eliminating the pathogen. This process is controlled by the chemotactic movement of chemokines induced as a primary host immune response. We have also addressed that chemokine expressions during infection are time-dependent and orchestrated in a systematic pattern that ultimately assists in generating a protective immune response. Taken together, this review provides a systematic understanding of the complexity of chemokines profiles during protozoan disease conditions and the rationale of targeting chemokines for the development of therapeutic strategies.
... With the use of the powerful tools available in mice, namely genetically modified mice, twophoton intravital microscopy of the brain, and genetically modified parasites, it has been established that key elements in ECM include the presence of parasites and infiltration of leukocytes in the brains of PbA-infected mice 44,45 . Although some studies have pointed to the role of macrophages 46 , monocytes 47 , neutrophils 48 , CD4 + T cells 49 , and natural killer cells 50 in ECM, others have shown that neither genetic nor antibody-mediated depletion of these cells mitigated disease [51][52][53][54] . However, the evidence for the role of CD8 + T cells is well established, as demonstrated in a multitude of depletion and effector function assays 44,45,49,51,52 . ...
Article
Full-text available
Cerebral malaria is the deadliest complication of malaria, a febrile infectious disease caused by Plasmodium parasite. Any of the five human Plasmodium species can cause disease, but, for unknown reasons, in approximately 2 million cases each year P. falciparum progresses to severe disease, ultimately resulting in half a million deaths. The majority of these deaths are in children under the age of five. Currently, there is no way to predict which child will progress to severe disease and there are no adjunctive therapies to halt the symptoms after onset. Herein, we discuss what is known about the disease mechanism of one form of severe malaria, cerebral malaria, and how we might exploit this understanding to rescue children in the throes of cerebral disease.
... MCP-1/CCL2 receptor CCR2. Ccr2-deficiency abrogated CCL2induced endothelial permeability in vitro (Stamatovic et al., 2003), but did not protect against ECM (Belnoue et al., 2003), indicating that other chemokines and/or additional mechanisms induce blood-brain barrier permeability. ...
Article
Full-text available
Cerebral malaria is among the major causes of malaria-associated mortality and effective adjunctive therapeutic strategies are currently lacking. Central pathophysiological processes involved in the development of cerebral malaria include an imbalance of pro- and anti-inflammatory responses to Plasmodium infection, endothelial cell activation, and loss of blood-brain barrier integrity. However, the sequence of events, which initiates these pathophysiological processes as well as the contribution of their complex interplay to the development of cerebral malaria remain incompletely understood. Several cytokines and chemokines have repeatedly been associated with cerebral malaria severity. Increased levels of these inflammatory mediators could account for the sequestration of leukocytes in the cerebral microvasculature present during cerebral malaria, thereby contributing to an amplification of local inflammation and promoting cerebral malaria pathogenesis. Herein, we highlight the current knowledge on the contribution of cytokines and chemokines to the pathogenesis of cerebral malaria with particular emphasis on their roles in endothelial activation and leukocyte recruitment, as well as their implication in the progression to blood-brain barrier permeability and neuroinflammation, in both human cerebral malaria and in the murine experimental cerebral malaria model. A better molecular understanding of these processes could provide the basis for evidence-based development of adjunct therapies and the definition of diagnostic markers of disease progression.
... Other chemokine receptors have also been implicated in the pathogenesis of malaria. CCR2 has been shown in murine experimental models to play little role in the development of CM (Belnoue et al. 2003a). CCR2 -/mice typically exhibit the inability of monocytes to traffic out of the bone marrow (Serbina and Pamer, 2006;Tsou et al. 2007;Jia et al. 2008;. ...
Article
Protists are a diverse collection of eukaryotic organisms that account for a significant global infection burden. Often, the immune responses mounted against these parasites cause excessive inflammation and therefore pathology in the host. Elucidating the mechanisms of both protective and harmful immune responses is complex, and often relies of the use of animal models. In any immune response, leucocyte trafficking to the site of infection, or inflammation, is paramount, and this involves the production of chemokines, small chemotactic cytokines of approximately 8-10 kDa in size, which bind to specific chemokine receptors to induce leucocyte movement. Herein, the scientific literature investigating the role of chemokines in the propagation of immune responses against key protist infections will be reviewed, focussing on Plasmodium species, Toxoplasma gondii, Leishmania species and Cryptosporidium species. Interestingly, many studies find that chemokines can in fact, promote parasite survival in the host, by drawing in leucocytes for spread and further replication. Recent developments in drug targeting against chemokine receptors highlights the need for further understanding of the role played by these proteins and their receptors in many different diseases.
Article
Full-text available
Objectives The immunologic events that build up to the fatal neurological stage of experimental cerebral malaria (ECM) are incompletely understood. Here, we dissected the immune cell behaviour that occurs in the central nervous system (CNS) when Plasmodium berghei-ANKA (PbA)-infected mice show only minor clinical signs. Methods We used a 2-photon intravital microscopy brain-imaging model to study the spatio-temporal context of early immunological events in situ during ECM. Results We found that early in the disease course, antigen-specific CD8+ T cells come in contact and arrest on the endothelium of post-capillary venules. CD8+ T cells typically adhered adjacent to or in the near vicinity of perivascular macrophages (PVM) that line the post-capillary venules. Closer examination revealed that CD8+ T cells crawled along the inner vessel wall towards PVM that lay on the abluminal side of large post-capillary venules. T cells typically localised at ‘activity hotspots’ in large post-capillary venules characterized by activated morphology and clustering of PVM, increased abutting of post-capillary venules by PVM as well as augmented monocyte accumulation. In the later stage of infection when mice exhibited neurological signs, intravascular CD8+ T cells increased in number and changed their behaviour, actively crawling along the endothelium and displaying frequent, short-term interactions with the inner vessel wall at hotspots. Conclusion Our study suggests an active interplay between the innate and adaptive arms of the immune system across the blood-brain barrier (BBB) in early ECM, which may be the initiating event in the inflammatory cascade leading to BBB alteration and neuropathology.
Chapter
Chemokine receptors, a family of G-protein-coupled receptors (GPCRs), bind in a specific manner to chemokines and elicit cellular responses. Their involvement in inflammatory diseases is predominant. Although the main function of chemokine receptors is enrolment of leukocytes at the site of inflammation, they are also widely explored as drug discovery targets. This is due to the fact that blockage of chemokine receptor may provide novel therapeutic interventions. This chapter discusses the various chemokine receptors, involvement of chemokine receptors in the pathogenesis of various diseases, and receptor-mediated strategies to tackle such afflictions.
Chapter
Breast cancer (BC) is the most common cause of death among women worldwide. Characterized by heterogeneous nature, treatment of breast cancer becomes very challenging considering its late detection. Causes for breast cancer and their treatment modalities have been well identified; still management of disease is an uphill task due to complex pathophysiology associated with it. Chemotherapy has remained the mainstay for treatment of BC, although with advent of novel targeted therapies, a paradigm shift is seen in treatment options available for BC. Targeted regimes toward receptors expressed on tumorous surfaces are developed that deploy antibodies and peptides for treating BC. Moreover, with avoidance of side effects of chemotherapy with concomitant annihilation of cancerous cells, it is very imperative to understand the underlying mechanisms of receptors governing the process. Therefore, with an attempt toward comprehensive understanding of the subject, this chapter explores some of the important receptors involved in breast cancer such as estrogen receptor, progesterone receptor, and human epidermal receptor-2. Special emphasis is given for the modulation of their signal transduction mechanism attaining desired goals. Various formulation aspects that are currently undertaken toward BC management are also discussed in brief.
Article
Full-text available
Malaria is a common and sometimes fatal disease caused by infection with Plasmodium parasites. Cerebral malaria (CM) is a most severe complication of infection with Plasmodium falciparum parasites which features a complex immunopathology that includes a prominent neuroinflammation. The experimental mouse model of cerebral malaria (ECM) induced by infection with Plasmodium berghei ANKA has been used abundantly to study the role of single genes, proteins and pathways in the pathogenesis of CM, including a possible contribution to neuroinflammation. In this review, we discuss the Plasmodium berghei ANKA infection model to study human CM, and we provide a summary of all host genetic effects (mapped loci, single genes) whose role in CM pathogenesis has been assessed in this model. Taken together, the reviewed studies document the many aspects of the immune system that are required for pathological inflammation in ECM, but also identify novel avenues for potential therapeutic intervention in CM and in diseases which feature neuroinflammation.
Article
Full-text available
We investigated the immune responses in mice lacking CCR2, CCR5, or macrophage inflammatory protein-1α (MIP-1α), a ligand for CCR5, in two situations: following T cell stimulation or after challenge with Leishmania donovani, an intracellular microbe whose control is dependent on a Th1 immune response. Mice deficient in CCR5, MIP-1α, or CCR2 had reduced IFN-γ responses following ligation of the TCR. Reduced IFN-γ responses following PMA and ionomycin were also observed in CD8+ T cells of CCR5−/− and CCR2−/− mice. During the early phases of infection, all three knockout mice had low Ag-specific IFN-γ responses. However, this reduced IFN-γ response was overcome during a state of persistent Ag stimulation (chronic infection), and was not associated with an adverse parasitologic outcome in any of the gene-targeted mouse strains. To the contrary, during the late phase of infection, an exaggerated Ag-specific IFN-γ response was evident in CCR5−/− and MIP-1α−/− mice, and this correlated with an enhanced control of parasite replication. Although granuloma formation was abnormal in each of the knockout mice, there was no correlation between the number or architecture of the granulomas and parasite burden. Collectively, these findings indicate an important role for CCR5, MIP-1α, and CCR2 in granulomatous inflammation, and that CCR5 and MIP-1α, possibly acting through CCR5, might play a deleterious role in the outcome of chronic L. donovani infection. Our data also suggest that there might be cross-talk between TCR and chemokine receptor signaling pathways.
Article
Full-text available
There is growing evidence that chemokines and their receptors regulate the movement and interaction of antigen-presenting cells such as dendritic cells (DCs) and T cells. We tested the hypothesis that the CC chemokine receptor (CCR)2 and CCR5 and the chemokine macrophage inflammatory protein (MIP)-1α, a ligand for CCR5, influence DC migration and localization. We found that deficiency of CCR2 but not CCR5 or MIP-1α led to distinct defects in DC biology. Langerhans cell (skin DC) density in CCR2-null mice was normal, and their ability to migrate into the dermis was intact; however, their migration to the draining lymph nodes was markedly impaired. CCR2-null mice had lower numbers of DCs in the spleen, and this was primarily due to a reduction in the CD8α1 T helper cell type 1 (Th1)-inducing subset of DCs. Additionally, there was a block in the Leishmania major infection–induced relocalization of splenic DCs from the marginal zone to the T cell areas. We propose that these DC defects, in conjunction with increased expression of B lymphocyte chemoattractant, a B cell–specific chemokine, may collectively contribute to the striking B cell outgrowth and Th2 cytokine–biased nonhealing phenotype that we observed in CCR2-deficient mice infected with L. major. This disease phenotype in mice with an L. major–resistant genetic background but lacking CCR2 is strikingly reminiscent of that observed typically in mice with an L. major–susceptible genetic background. Thus, CCR2 is an important determinant of not only DC migration and localization but also the development of protective cell-mediated immune responses to L. major.
Article
Full-text available
CC chemokine receptor 2 (CCR2) is a prominent receptor for the monocyte chemoattractant protein (MCP) group of CC chemokines. Mice generated by gene targeting to lack CCR2 exhibit normal leukocyte rolling but have a pronounced defect in MCP-1-induced leukocyte firm adhesion to microvascular endothelium and reduced leukocyte extravasation. Constitutive macrophage trafficking into the peritoneal cavity was not significantly different between CCR2-deficient and wild-type mice. However, after intraperitoneal thioglycollate injection, the number of peritoneal macrophages in CCR2-deficient mice did not rise above basal levels, whereas in wild-type mice the number of macrophages at 36 h was ≈3.5 times the basal level. The CCR2-deficient mice showed enhanced early accumulation and delayed clearance of neutrophils and eosinophils. However, by 5 days neutrophils and eosinophils in both CCR2-deficient and wild-type mice had returned to near basal levels, indicating that resolution of this inflammatory response can occur in the absence of macrophage influx and CCR2-mediated activation of the resident peritoneal macrophages. After intravenous injection with yeast β-glucan, wild-type mice formed numerous large, well-defined granulomas throughout the liver parenchyma, whereas CCR2-deficient mice had much fewer and smaller granulomas. These results demonstrate that CCR2 is a major regulator of induced macrophage trafficking in vivo.
Article
IFN- has been implicated in the pathogenesis of experimental cerebral malaria (ECM). We have used mice lacking the chain of the IFN- receptor (KO mice) to define its role in the pathogenesis of ECM. Infected KO mice did not develop ECM and showed no leukocyte or parasite sequestration in the brain, and no hemorrhages. The resistance of KO mice to ECM was associated with the absence of any increases of TNF- and ICAM-1 proteins in the brain, which are both essential for ECM. Wild-type (WT) mice which do not develop ECM, despite increased local production of TNF- protein, showed no leukocyte accumulation in the brain and this was correlated with the absence of ICAM-1 protein from brain microvessels. KO mice infected with 106 parasitized erythrocytes (PE) of Plasmodium berghei ANKA (PbA) did not develop ECM, but they had high parasitemia and died earlier than WT mice which did not develop ECM. However, KO mice did not develop higher parasitemia than WT mice when both groups were infected with a lower dose (5×105 PE) of PbA-infected red blood cells. This indicates that different doses of PE may trigger different IFN- responses and that there may be a threshold concentration for protection against parasitemia.
Article
Guidelines for submitting commentsPolicy: Comments that contribute to the discussion of the article will be posted within approximately three business days. We do not accept anonymous comments. Please include your email address; the address will not be displayed in the posted comment. Cell Press Editors will screen the comments to ensure that they are relevant and appropriate but comments will not be edited. The ultimate decision on publication of an online comment is at the Editors' discretion. Formatting: Please include a title for the comment and your affiliation. Note that symbols (e.g. Greek letters) may not transmit properly in this form due to potential software compatibility issues. Please spell out the words in place of the symbols (e.g. replace “α” with “alpha”). Comments should be no more than 8,000 characters (including spaces ) in length. References may be included when necessary but should be kept to a minimum. Be careful if copying and pasting from a Word document. Smart quotes can cause problems in the form. If you experience difficulties, please convert to a plain text file and then copy and paste into the form.
Article
CBA/T6 and Balb/c mice inoculated with Plasmodium berghei ANKA strain (PbA) died from cerebral malaria 6-8 days post-inoculation. DBA/2J mice similarly inoculated developed a non-fatal cerebral malaria, with mild temporary cerebral symptoms, and died between days 15 and 22 from other malaria-related complications. When inoculated with P. berghei K173 (Pb) these mouse strains did not develop a cerebral malaria but died between days 15 and 22 from other malaria-related complications. These mouse strain/parasite strain combinations allow for detailed examination of factors critical in the pathology of murine cerebral malaria. Monastral Blue, a colloid dye, when injected intravascularly between days 0 and 2 into PbA-inoculated CBA (PbA-CBA) or Balb/c (PbA-Balb/c) mice prevented death from cerebral malaria. There was no evidence of increased vascular permeability at this stage. When Monastral Blue was injected between days 5 and 8, there was increased vascular permeability in the kidney, liver, lung, spleen and brain of PbA-CBA and PbA-Balb/c mice. Injection of Monastral Blue into these animals at this time also precipitated cerebral symptoms and death, but not in Pb-infected mice. Endothelial and mononuclear cells phagocytosed, and were coated with, the Monastral Blue particles when the dye was injected between days 5 and 8 into PbA-CBA and PbA-Balb/c mice. Control, uninfected mice did not demonstrate either of these features. Pb-infected mice only demonstrated coated mononuclear cells. Mononuclear cell attachment to the endothelium, increased vascular permeability and increased association of Monastral Blue particles with monocytes and endothelial cells were correlated with cerebral symptoms and death. Monastral Blue is thus a useful agent for studying the roles of mononuclear cells and endothelium in murine cerebral malaria.
Article
For investigation of the pathogenesis of cerebral malaria, immediate postmortem samples from brain and other tissues of patients dying with Plasmodium falciparum malaria, with (CM) or without (NCM) cerebral malaria, were processed for electron microscopy. Counts of parasitized erythrocytes (PRBCs) in cerebral and other vessels showed that the proportion of PRBCs was higher in CM than in NCM, and also that the proportion of PRBCs was higher in the brain than in other organs examined in both CM and NCM. Cerebral vessels from CM patients were more tightly packed with RBCs than those from NCM patients, but there was no significant difference in the amount or degree of endothelial damage or numbers of vessels with endothelial pseudopodia. Fibrillar (fibrin) deposits were present in a small proportion of vessels, but no thrombosis was present. There was neither acute nor chronic inflammation, and leukocytes were absent within or outside cerebral vessels. There was no immune complex deposition in cerebral vessels. Parasites in cerebral vessels were mainly trophozoites or schizonts. Occasional RBC remnants following parasite release were seen. Some parasites were degenerate, resembling crisis forms. PRBCs adhered to endothelium via surface knobs. It is concluded that there is no evidence for an inflammatory or immune pathogenesis for human cerebral malaria and that the clinical effects probably relate to anoxia and the metabolic activities of the parasites.
Article
A panel of hybridomas secreting monoclonal antibodies (mAb) reacting with antigens on mouse eosinophils and neutrophils has been selected. It is shown that four mAb bind preferentially to eosinophils, recognizing antigens expressed between 5 and 25 times more densely on these cells than on neutrophils. One mAb reacts preferentially with neutrophils, binding an antigen about 12 times more common on these cells than on eosinophils. Four mAb are shown to react with both eosinophils and neutrophils, and also macrophages. These mAb confirm the presence of different differentiation antigens on both eosinophils and neutrophils, and show that the myeloid series express antigens not expressed on lymphocytes.
Article
Plasmodium berghei ANKA was tested for its usefulness as a model for cerebral malaria in inbred mouse strains A, A2G, A/J, C57L, SJL/J and SWR. A suitable model was obtained in A or A/J strain mice. Mortality was 100% after five to eight days with brain haemorrhages occurring terminally. The histopathology is described. The model was stable after six blood passages at 5- to 7-day intervals. Chloroquine abolished the haemorrhages and no intercurrent viral or blood protozoal infections were detected.