Article

Cerebral Malaria in Mice: Demonstration of Cytoadherence of Infected Red Blood Cells and Microrheologic Correlates

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract

To understand the microcirculatory events during cerebral malaria, we have studied the lethal strain of rodent Plasmodia, Plasmodium yoelii 17XL, originally described by Yoeli and Hargreaves in 1974. The virulence of P. yoelii 17XL is caused by intravascular sequestration of infected red blood cells (IRBCs), especially in the brain vessels and capillaries. This mouse model resembles human P. falciparum infection more closely than P. berghei ANKA infection since it shows little, if any, inflammation of the brain. In vivo microcirculatory studies on cytoadherence of IRBCs were performed using the cremaster muscle preparation, which is an easily accessible vasculature for intravital observations. Ex vivo assay of cytoadherence was carried out in the artificially perfused mesocecum preparation of the rat. The results in either preparation demonstrated cytoadherence of IRBCs that was restricted to postcapillary venules. Furthermore, the in vivo measurements showed the prevalence of cytoadherence in small-diameter (< 40 microns) venules in accordance with the local wall shear rates. The parasitized animals demonstrated significantly reduced red blood cell velocities and wall shear rates in the small-diameter postcapillary venules of the cremaster. The relationship between cytoadherence and venular wall shear rates was also reflected in the inverse correlation between the number of adhered cells and the venular diameter in the ex vivo mesocecum preparation. In the ex vivo preparation, cytoadherence of IRBCs was accompanied by a higher peripheral resistance. Transmission electron microscopy of the cremaster muscle and brain tissues showed a tight association of IRBCs with the endothelium of small venules. These observations demonstrate that cytoadherence of P. yoelii 17XL-infected mouse red blood cells is very similar to that of P. falciparum-infected cells. Thus, this model should allow a detailed analysis of the molecular mechanisms involved in the generation of cerebral malaria by cytoadherence of the infected red blood cells to the vascular endothelium.

No full-text available

Request Full-text Paper PDF

To read the full-text of this research,
you can request a copy directly from the authors.

... Importantly, studies in vivo and ex vivo demonstrated cytoadherence of infected RBC in the postcapillary venules, decreased red blood cell velocities, and increased shear stress. 11 This model is different than the P. berghei ANKA model in which the cerebral malaria seems to have an inflammatory pathogenesis, 12 unlike human cerebral malaria. 2 To understand further the pathogenesis of cerebral malaria in the P. yoelii model, we studied the expression of two ligands, ICAM-1 and VCAM-1, thought to be involved in adhesion of infected red blood cells. Our results suggest that up-regulation of ICAM-1 is a distinctive feature of P. yoelii cerebral malaria. ...
... There is an accumulation of infected RBCs in the brain, in the absence of an influx of mononuclear cells. 11 Importantly, in the P. yoelii 17XL model, we observed cytoadherence in vivo, mainly of infected RBCs, and reduced blood flow. 11 Although P. yoelii-infected RBCs do not display surface knobs, close adherence of infected RBCs to brain endothelium was observed by electron microscopy. ...
... 11 Importantly, in the P. yoelii 17XL model, we observed cytoadherence in vivo, mainly of infected RBCs, and reduced blood flow. 11 Although P. yoelii-infected RBCs do not display surface knobs, close adherence of infected RBCs to brain endothelium was observed by electron microscopy. 11 Like P. falciparum, P. yoelii 17XL can invade both mature and immature RBCs. ...
Article
Previous studies demonstrated that Plasmodium yoelii 17XL, a lethal strain of rodent malaria, causes a syndrome in SW mice that resembles human cerebral malaria. The mouse brain pathology is characterized by cy- toadherence of parasitized erythrocytes. Here, the possible mechanisms mediating cerebral malaria in this model were studied and the results were compared with a nonlethal strain of this parasite, P. yoelii 17XNL (nonlethal), which does not cause cerebral malaria. Immunostaining for intercellular adhesion molecule-1 (ICAM-1) revealed an increase in expression of this protein in the small venules and capillaries of the brains of infected mice that increased with time after infection. Staining was more pronounced during the lethal infection than the nonlethal infection. Some staining with monoclonal antibody to vascular cell adhesion molecule-1 was also observed, but it was quantitatively less than ICAM-1 staining and was limited to larger venules. During the lethal infection, levels of tumor necrosis factor- a (TNF-a) increased rapidly, peaking on day 4. In contrast, mice infected with nonlethal P. yoelii had a slower serum TNF-a response that peaked on day 10, prior to the maximum parasitemia. In addition, mice with a targeted disruption of the TNF-a gene (TNF-a2/2 mice) were infected with the lethal and nonlethal strains of P. yoelii 17X. The TNF-a2/2 mice infected with the nonlethal parasite had significantly higher levels of parasitemia than controls, whereas TNF-a2/2 mice infected with the lethal strain had slightly higher levels of infected erythrocytes but were equally susceptible to death from this infection. Thus, TNF-a does not appear to be essential in mediating death. These results demonstrate that P. yoelii 17XL infection has features in common with human cerebral malaria and suggest that this model may be useful in testing strategies to alleviate this syndrome. Cerebral malaria refers to the severe complications of fal- ciparum malaria in which vascular plugging of Plasmodium falciparum-infected red blood cells (RBCs) in the brain can lead to coma and death, often in children. 1,2 Cytoadherence
... Rodents-Mouse models of CM are the most commonly used in vivo experimental model [39]. Several mouse models exist in which infection is induced with Plasmodium infected RBC including P. yoelii 17XL [69], PbA [70][71][72] P. berghei NK65 [73] and PbK173 [74]. Each model exhibits specific characteristics, P. yoelii for instance infects preferentially reticulocytes while the others infect mature RBC. ...
... In vitro assays using mouse brain MVEC have provided direct in vitro evidence for cytoadherence of RBC from P. berghei ANKA (PbA) and P. berghei K173 (PbK173)infected mice on brain endothelial cells [83]. PRBC infected with P. yoelii 17XL also adhere to mouse brain MVEC [70]. ...
... Both CD4+ and CD8+ T cells have been implicated in HCM and ECM pathogenesis [177][178][179] and athymic nude mice do not develop CM [179]. Helper T lymphocytes are also involved in the pathogenesis of CM, contributing to the development of cerebral lesions with microvessel plugging and haemorrhages [70,179]. Depletion of CD4+ and CD8+ T cells reduced parasite biomass, and mice did not develop CM [44]. ...
Article
Full-text available
Human cerebral malaria is a severe and often lethal complication of Plasmodium falciparum infection. Complex host and parasite interactions should the precise mechanisms involved in the onset of this neuropathology. Adhesion of parasitised red blood cells and host cells to endothelial cells lead to profound endothelial alterations that trigger immunopathological changes, varying degrees of brain oedema and can compromise cerebral blood flow, cause cranial nerve dysfunction and hypoxia. Study of the cerebral pathology in human patients is limited to clinical and genetic field studies in endemic areas, thus cerebral malaria (CM) research relies heavily on experimental models. The availability of malaria models allows study from the inoculation of Plasmodium to the onset of disease and permit invasive experiments. Here, we discuss some aspects of our current understanding of CM, the experimental models available and some important recent findings extrapolated from these models.
... The cytoadherence of infected red blood cells (IRBCs) to the postcapillary venules is the major cause of IRBC sequestration and vessel blockage in the cerebral form of human malaria. In both human cerebral malaria caused by P. falciparum and the Plasmodium yoelii 17XL-infected rodent model of malaria, the sequestration of IRBCs in the brain vessels is secondary to the cytoadherence of IRBCs to the postcapillary venules [14]. In this study, we analysed the alterations in immunomodulator gene expression in brain samples of P. yoelii 17XL-infected mice using cDNA microarray screening, coupled with analysis of temporal expression patterns of RANTES and its corresponding receptors, CCR1, CCR3 and CCR5, in brain samples and plasma of P. yoelii 17XL-infected mice to identify and characterize the role of these immunomodulators during rodent malaria. ...
... Mice were injected intraperitoneally with P. yoelii 17XL parasitized blood, kindly provided by Dr. Christine Olver (Department of Pathology, Colorado State University, USA). This rodent malaria strain causes a syndrome that resembles human malaria, char acterized by fever, spleno-and hepatomegaly by day eight post-infection [14,15]. Parasitaemia was determined in a total count of 300 to 500 red blood cells (RBCs) on Wright-Giemsa-stained (Sigma Diagnostics, USA) thin blood smears. ...
... All the mice infected with P. yoelii 17XL parasites developed malaria-related-symptoms, which included the appearance of ruffled fur and shivering at peak parasitaemia by day eight post-infection ( Figure 1). Examination of the viscera of dissected mice confirmed spleno-and hepatomegaly at peak parasitaemia, concordant with reported P. yoelii 17XL malaria infections [14,15]. None of the control or uninfected mice showed any of these signs. ...
Article
Full-text available
Abstract Background Malaria afflicts 300–500 million people causing over 1 million deaths globally per year. The immunopathogenesis of malaria is mediated partly by co mplex cellular and immunomodulator interactions involving co-regulators such as cytokines and adhesion molecules. However, the role of chemokines and their receptors in malaria immunopathology remains unclear. RANTES (Regulated on Activation Normal T-Cell Expressed and Secreted) is a chemokine involved in the generation of inflammatory infiltrates. Recent studies indicate that the degradation of cell-cell junctions, blood-brain barrier dysfunction, recruitment of leukocytes and Plasmodium -infected erythrocytes into and occlusion of microvessels relevant to malaria pathogenesis are associated with RANTES expression. Additionally, activated lymphocytes, platelets and endothelial cells release large quantities of RANTES, thus suggesting a unique role for RANTES in the generation and maintenance of the malaria-induced inflammatory response. The hypothesis of this study is that RANTES and its corresponding receptors (CCR1, CCR3 and CCR5) modulate malaria immunopathogenesis. A murine malaria model was utilized to evaluate the role of this chemokine and its receptors in malaria. Methods The alterations in immunomodulator gene expression in brains of Plasmodium yoelii 17XL-infected mice was analysed using cDNA microarray screening, followed by a temporal comparison of mRNA and protein expression of RANTES and its corresponding receptors by qRT-PCR and Western blot analysis, respectively. Plasma RANTES levels was determined by ELISA and ultrastructural studies of brain sections from infected and uninfected mice was conducted. Results RANTES (p < 0.002), CCR1 (p < 0.036), CCR3 (p < 0.033), and CCR5 (p < 0.026) mRNA were significantly upregulated at peak parasitaemia and remained high thereafter in the experimental mouse model. RANTES protein in the brain of infected mice was upregulated (p < 0.034) compared with controls. RANTES plasma levels were significantly upregulated; two to three fold in infected mice compared with controls (p < 0.026). Some d istal microvascular endothelium in infected cerebellum appeared degraded, but remained intact in controls. Conclusion The upregulation of RANTES, CCR1, CCR3, and CCR5 mRNA, and RANTES protein mediate inflammation and cellular degradation in the cerebellum during P. yoelii 17XL malaria.
... respects including cytoadherence of parasitized red blood cells (PRBCs) in the brain and pathological symptoms. 7,8 Whether CM represents an inflammatory pathogenesis was not clarified by studying P. yoelii infection in susceptible mice. It remains debatable whether human CM presents an inflammatory pathogenesis. ...
... 36 No infiltration of lymphocytes and parasitized erythrocytes or endothelial damage was seen in the brains of mice that were infected but received no IL-2 treatment, or in mice that were given IL-2 only. The histological changes seen in the brain of IL-2-treated and infected DBA/2 mice closely resemble those described in susceptible mice (see Figure 3 in this study and Yoelii and Hargreaves 7 and Kaul et al 8 ). ...
Article
Full-text available
In this study, we report that infection with Plasmo-dium yoelii 17XL, a lethal strain of rodent malaria, does not result in death in the DBA/2 strain of mice. In contrast to BALB/c mice, DBA/2 mice developed significantly less parasitemia and never manifested symptoms of cerebral malaria (CM) on infection with this parasite. Moreover, the histological changes evident in the brain of susceptible BALB/c were absent in DBA/2 mice. Interestingly, the resistant DBA/2 mice when treated with recombinant interleukin (IL)-2, were found to develop CM symptoms and the infection became fatal by 6 to 8 days after infection. This condition was associated with an augmented interfer-on-␥ and nitric oxide production. Unexpectedly, IL-10 levels were also elevated in IL-2-treated DBA/2 mice during late stage of infection (at day 6 of infection) whereas the inverse relationship between IL-10 and interferon-␥ or nitric oxide was maintained in the early stage of infection (at day 3 after infection). The level of tumor necrosis factor-␣ production was moderately increased in the late phase of infection in these mice. Histology of brain from IL-2-treated mice demonstrated the presence of parasitized erythro-cytes and infiltration of lymphocytes in cerebral vessels , and also displayed some signs of endothelial degeneration. Confocal microscopical studies demonstrated preferential accumulation of ␥␦ T cells in the cerebral vessels of IL-2-treated and-infected mice but not in mice treated with IL-2 alone. The cells recruited in the brain were activated because they demonstrated expression of CD25 (IL-2R) and CD54 (inter-cellular adhesion molecule 1) molecules. Administration of anti-␥␦ mAb prevented development of CM in IL-2-treated mice until day 18 after infection whereas mice treated with control antibody showed CM symptoms by day 6 after infection. The information concerning creating pathological sequelae and death in an otherwise resistant mouse strain provides an interesting focus for the burden of pathological attributes on death in an infectious disease. (Am J Pathol 2001, 158:163-172) The pathogenesis of cerebral malaria (CM) has been a subject of considerable interest, and it remains a major cause of death associated with severe Plasmodium falci-parum infection in many tropical countries. 1 The early events that lead to human CM are difficult to study experimentally because of ethical constraints and the limitations of postmortem materials. Because of these difficulties , investigators turned to murine models to investigate mechanisms of CM pathogenesis. Considerable effort has been devoted to study the cerebral pathogenesis in the murine models involving Plasmodium berghei ANKA and Plasmodium yoelii 17XL, however, there are many aspects of this pathogenesis that are still poorly understood. 2 Previous studies in the P. berghei ANKA murine model have indicated that CM might be a lymphocyte-mediated disease, in which CD4ϩ, and CD8ϩ T cells have been postulated to play a role. 3-5 It was recently demonstrated that on treatment with anti-␥␦ T cell antibody, the susceptible mice failed to develop CM after P. berghei ANKA infection indicating some pathological role of ␥␦ T cells. 6 However, the extent of T cell activation that is required for the development of CM and whether actual T cell infiltration occurs at the site of brain has not been known. Morphological and biochemical studies have so far not recognized a focal or global determi-nant(s) for CM pathogenesis or a clear relationship between brain lesion and death. P. yoelii 17XL regularly induces a CM syndrome in susceptible mice that parallels human disease in several Supported by institutional funding to Unité de Recherche 399, INSERM.
... The reason this response was not sustained and instead turned into a vasoconstriction is unknown, but it may be related to low NO bioavailability. 34 Vasoconstriction and vascular collapse have not been described in previous intravital studies of the skin and muscle microcirculation in rodent models of severe malaria, [22][23][24] although in the P. yoelii model major decreases in blood flow were shown. 22,23 However, these models and the sites of observations differ significantly from the present study and cannot be directly compared. ...
... 34 Vasoconstriction and vascular collapse have not been described in previous intravital studies of the skin and muscle microcirculation in rodent models of severe malaria, [22][23][24] although in the P. yoelii model major decreases in blood flow were shown. 22,23 However, these models and the sites of observations differ significantly from the present study and cannot be directly compared. ...
Article
Brain hemodynamics in cerebral malaria (CM) is poorly understood, with apparently conflicting data showing microcirculatory hypoperfusion and normal or even increased blood flow in large arteries. Using intravital microscopy to assess the pial microvasculature through a closed cranial window in the murine model of CM by Plasmodium berghei ANKA, we show that murine CM is associated with marked decreases (mean: 60%) of pial arteriolar blood flow attributable to vasoconstriction and decreased blood velocity. Leukocyte sequestration further decreased perfusion by narrowing luminal diameters in the affected vessels and blocking capillaries. Remarkably, vascular collapse at various degrees was observed in 44% of mice with CM, which also presented more severe vasoconstriction. Coadministration of artemether and nimodipine, a calcium channel blocker used to treat postsubarachnoid hemorrhage vasospasm, to mice presenting CM markedly increased survival compared with artemether plus vehicle only. Administration of nimodipine induced vasodilation and increased pial blood flow. We conclude that vasoconstriction and vascular collapse play a role in murine CM pathogenesis and nimodipine holds potential as adjunctive therapy for CM.
... The outbred Swiss mouse has long been used in experimental malaria studies, but reports of its use in CM studies are scarce (Gupta et al. 1988;Kaul et al. 1994;Franke-Fayard et al. 2005) and description of PbA infection in this mouse stock lacks details on the course of infection, CM incidence and histopathological alterations. In this study, we describe the occurrence of CM in the outbred Swiss Webster mouse and characterize the associated multiorgan pathology by histology. ...
... This newly characterized mouse-parasite combination adds to a number of existing murine models of CM, mostly using the PbA strain in several inbred mouse strains, such as the CBA and the C57Bl ⁄ 6, and also others using different rodent plasmodial species (de Kossodo & Grau 1993;de Souza & Riley 2002). This is the case, for instance, for the infection of Balb ⁄ c or Swiss Webster mice by Plasmodium yoelii, claimed to be models in which pRBC sequestration occurs (Kaul et al. 1994). In addition, extensive characterization of P. berghei infection in the Swiss Webster mouse was performed in the early works of Jerusalem and Desowitz (Van Zon et al. 1978;Jerusalem et al. 1983). ...
Article
Plasmodium berghei ANKA (PbA) infection in susceptible inbred mouse strains is the most commonly used experimental model to study pathogenesis of cerebral malaria (CM). Indeed, many concepts on mechanisms related to this complication have arisen from works using this model. Although inbred strains present several advantages and are indicated for most studies, the use of outbred models can show unique usefulness in a number of approaches such as fine post-quantitative trait loci mapping and discovery of genes relevant to CM susceptibility or resistance, as well as pharmacological and vaccine studies. Here we describe the features of PbA infection and CM incidence, and characterize the associated multiorgan pathology in the outbred Swiss Webster mouse. This model showed a sizeable (62.7%) and reproducible incidence of CM demonstrated by clinical signs and histopathological changes in brain (microhaemorrhages, oedema and vessel plugging by mononuclear cells). Major pathological changes were also observed in lungs, liver, thymus and spleen, analogous to those observed in inbred strains. Parasitaemia levels were associated with the risk of CM development, the risk being significantly higher in mice showing higher values of parasitaemia on days 6-7 of infection. This outbred CM model is then suitable for genetic, vaccine and drug studies targeting this malaria complication.
... respects including cytoadherence of parasitized red blood cells (PRBCs) in the brain and pathological symptoms. 7,8 Whether CM represents an inflammatory pathogenesis was not clarified by studying P. yoelii infection in susceptible mice. It remains debatable whether human CM presents an inflammatory pathogenesis. ...
... 36 No infiltration of lymphocytes and parasitized erythrocytes or endothelial damage was seen in the brains of mice that were infected but received no IL-2 treatment, or in mice that were given IL-2 only. The histological changes seen in the brain of IL-2-treated and infected DBA/2 mice closely resemble those described in susceptible mice (see Figure 3 in this study and Yoelii and Hargreaves 7 and Kaul et al 8 ). ...
Article
In this study, we report that infection with Plasmodium yoelii 17XL, a lethal strain of rodent malaria, does not result in death in the DBA/2 strain of mice. In contrast to BALB/c mice, DBA/2 mice developed significantly less parasitemia and never manifested symptoms of cerebral malaria (CM) on infection with this parasite. Moreover, the histological changes evident in the brain of susceptible BALB/c were absent in DBA/2 mice. Interestingly, the resistant DBA/2 mice when treated with recombinant interleukin (IL)-2, were found to develop CM symptoms and the infection became fatal by 6 to 8 days after infection. This condition was associated with an augmented interferon-gamma and nitric oxide production. Unexpectedly, IL-10 levels were also elevated in IL-2-treated DBA/2 mice during late stage of infection (at day 6 of infection) whereas the inverse relationship between IL-10 and interferon-gamma or nitric oxide was maintained in the early stage of infection (at day 3 after infection). The level of tumor necrosis factor-alpha production was moderately increased in the late phase of infection in these mice. Histology of brain from IL-2-treated mice demonstrated the presence of parasitized erythrocytes and infiltration of lymphocytes in cerebral vessels, and also displayed some signs of endothelial degeneration. Confocal microscopical studies demonstrated preferential accumulation of gammadelta T cells in the cerebral vessels of IL-2-treated and -infected mice but not in mice treated with IL-2 alone. The cells recruited in the brain were activated because they demonstrated expression of CD25 (IL-2R) and CD54 (intercellular adhesion molecule 1) molecules. Administration of anti-gammadelta mAb prevented development of CM in IL-2-treated mice until day 18 after infection whereas mice treated with control antibody showed CM symptoms by day 6 after infection. The information concerning creating pathological sequelae and death in an otherwise resistant mouse strain provides an interesting focus for the burden of pathological attributes on death in an infectious disease.
... Brain involvement similar to cerebral malaria does occur in related rodent species (P. berghei, Rest (1982) which also sequesters in other organs (Alger 1963), and in P. yoelii, Yoeli & Hargreaves 1974; Kaul et al. 1994), but this appears to be only after adaptation to the novel host (mice) through serial passage. In addition to its similarities with P. falciparum, the P. chabaudi system is attractive as a model because of the availability of diverse parasite strains cryopreserved shortly after they were derived from the wild (Beale et al. 1978), the availability of host genotypes with well-characterized genetics, the ability to measure virulence, multiplication rate, gametocyte production, transmission to mosquitoes, cytoadherence and antigenic variation in vivo, and the ability to manipulate immunity and other factors that moderate virulence. ...
... (a) Cytoadherence Cytoadherence refers to a group of related phenomena in which parasite-infected cells adhere to other host cells during the last half of the parasite's replication cycle: these host cells include endothelial cells in the post-capillary venules ('sequestering'), uninfected red blood cells ('rosetting'), infected red blood cells ('autoagglutination' or 'clumping') and leucocytes. In isolation or by interacting (Handunetti et al. 1992; Ho & White 1999), and perhaps exacerbated by reduced red cell deformability (Dondorp et al. 1997), these cytoadherence phenotypes lead to a slowing and obstruction of blood flow in the post-capillary venules (MacPherson et al. 1985; RaventosSuarez et al. 1985; Kaul et al. 1991 Kaul et al. , 1994 Nash et al. 1992 ) and lead to hypoxia and nitric oxide-related pathology (Clark & Rockett 1996; Dondorp et al. 1998). Together, these can lead to coma, organ failure and death. ...
Article
Malaria parasites cause much morbidity and mortality to their human hosts. From our evolutionary perspective, this is because virulence is positively associated with parasite transmission rate. Natural selection therefore drives virulence upwards, but only to the point where the cost to transmission caused by host death begins to outweigh the transmission benefits. In this review, we summarize data from the laboratory rodent malaria model, Plasmodium chabaudi, and field data on the human malaria parasite, P. falciparum, in relation to this virulence trade-off hypothesis. The data from both species show strong positive correlations between asexual multiplication, transmission rate, infection length, morbidity and mortality, and therefore support the underlying assumptions of the hypothesis. Moreover, the P. falciparum data show that expected total lifetime transmission of the parasite is maximized in young children in whom the fitness cost of host mortality balances the fitness benefits of higher transmission rates and slower clearance rates, thus exhibiting the hypothesized virulence trade-off. This evolutionary explanation of virulence appears to accord well with the clinical and molecular explanations of pathogenesis that involve cytoadherence, red cell invasion and immune evasion, although direct evidence of the fitness advantages of these mechanisms is scarce. One implication of this evolutionary view of virulence is that parasite populations are expected to evolve new levels of virulence in response to medical interventions such as vaccines and drugs.
... In mice infected with P. yoelii 17XNL strain, parasitemias reach a peak of 30 -40% by about 20 days, and mice generally survive by clearing infection (42)(43)(44). In contrast, mice infected with the sequestering, lethal P. yoelii 17XL strain die at 6 -10 days postinfection (pi), exhibiting peak parasitemias of 60 -80% (pi) (45)(46)(47). ...
Article
Full-text available
In malaria, CD36 plays several roles including mediating parasite sequestration to host organs, phagocytic clearance of parasites and regulation of immunity. While the functions of CD36 in parasite sequestration and phagocytosis have been clearly defined, less is known about its role in malaria immunity. Here, to understand the function of CD36 in malaria immunity, we studied parasite growth, innate and adaptive immune responses, and host survival in WT and Cd36-/-mice infected with a nonlethal strain of Plasmodium yoelii. Compared to Cd36-/-mice, WT mice had lower parasitemias and were resistant to death. At early but not at later stages of infection, WT mice had higher circulatory pro-inflammatory cytokines and lower anti-inflammatory cytokines than Cd36-/-mice. WT mice showed higher frequencies of pro-inflammatory cytokine-producing and lower frequencies of anti-inflammatory cytokine-producing DCs and NK cells than Cd36-/-mice. Cytokines produced by co-cultures of DCs from infected mice and OT-II T cells reflected CD36-dependent DC function. WT mice also showed increased Th1 and reduced Th2 responses than Cd36-/-mice, mainly at early stages of infection. Furthermore, in infected WT mice, macrophages and neutrophils expressed higher levels of phagocytic receptors and showed enhanced phagocytosis of parasite-infected erythrocytes than those in Cd36-/-mice, in an IFN-γ-dependent manner. However, there were no differences in malaria-induced humoral responses between WT and Cd36-/-mice. Overall, the results show that CD36 plays a significant role in controlling parasite burden by contributing to pro-inflammatory cytokine responses by DCs and NK cells, Th1 development, phagocytic receptor expression, and phagocytic activity.
... Therefore, the analysis of the interaction of the parasitized erythrocytes with host endothelium is practically restricted to the examination of postmortem samples. Rodent malaria models are useful for the study of many aspects of the disease, but the pathology of murine plasmodium infections is different from that of human cerebral malaria, and sequestration of parasitized erythrocytes to the brain vasculature has not been observed in the models used so far, except for Plasmodium yoelii XL, where infection was not associated with inflammatory reactions in the brain (9,10). An alternative is to investigate the adhesion of isolated P. fakiparum-infected erythrocytes to cultured cell lines in vitro. ...
Article
Cerebral malaria is a fatal complication of infection by Plasmodium falciparum in man. The neurological symptoms that characterize this form of malarial disease are accompanied by the adhesion of infected erythrocytes to the vasculature of the brain. To study this phenomenon in vivo, an acute phase severe combined immunodeficiency (SCID) mouse model was developed in which sequestration of P. falciparum-infected human erythrocytes took place. During acute cerebral malaria in humans, the expression of intercellular adhesion molecule-1 (ICAM-1) is induced in vascular endothelium by inflammatory reactions. Acute phase ICAM-1 expression can also be obtained in SCID mice. The endothelium of the midbrain region was the most responsive to such inflammatory stimulus. It is noteworthy that the reticular formation in the midbrain controls the level of consciousness, and loss of consciousness is a symptom of cerebral malaria. We found that infected human erythrocytes were retained 24 times more than normal erythrocytes in ICAM-1-positive mouse brain. Sequestration to the brain was reduced by anti-ICAM-1 antibodies. These in vivo results were confirmed by the binding of P. falciparum-infected erythrocytes to the ICAM-1-positive endothelium in tissue sections of mouse brain. We conclude that the SCID mouse serves as a versatile in vivo model that allows the study of P. falciparum-infected erythrocyte adhesion as it occurs in human cerebral malaria. Upregulation of ICAM-1 expression in the region of the midbrain correlates with increased retention of malaria-infected erythrocytes and with the symptoms of cerebral malaria.
... Our data do not support the assertion that elevated vcam-1 corresponds to ECM, as we observed slightly higher vcam-1 mRNA levels in ⌬hmgb2 ANKA-infected mice at d6 postinfection than in WT P. berghei ANKA-infected C57BL/6 mice, suggesting an uncertain or perhaps protective role for this adhesion molecule in our model. In concordance with our observation of a predominant role for ICAM-1 compared to VCAM-1 in malaria pathogenesis, infusion of an anti-ICAM-1 but not an anti-VCAM-1 monoclonal antibody prevented cytoadherence of infected erythrocytes in a P. yoelii model of ECM (65), in addition to in vivo evidence for the role of ICAM-1 in the sequestration of infected red blood cells in a mouse model of lethal malaria (66). In contrast, the transcript level of the cytoprotective heme oxygenase gene (hmox-1) was markedly increased at d6 in ⌬hmgb2 ANKAinfected C57BL/6 mice and was thus consistent with the literature (9)(10)(11). ...
Article
Full-text available
Eukaryotic high-mobility-group-box (HMGB) proteins are nuclear factors involved in chromatin remodeling and transcription regulation. When released into the extracellular milieu, HMGB1 acts as a proinflammatory cytokine that plays a central role in the pathogenesis of several immune-mediated inflammatory diseases. We found that the Plasmodium genome encodes two genuine HMGB factors, Plasmodium HMGB1 and HMGB2, that encompass, like their human counterparts, a proinflammatory domain. Given that these proteins are released from parasitized red blood cells, we then hypothesized that Plasmodium HMGB might contribute to the pathogenesis of experimental cerebral malaria (ECM), a lethal neuroinflammatory syndrome that develops in C57BL/6 (susceptible) mice infected with Plasmodium berghei ANKA and that in many aspects resembles human cerebral malaria elicited by P. falciparum infection. The pathogenesis of experimental cerebral malaria was suppressed in C57BL/6 mice infected with P. berghei ANKA lacking the hmgb2 gene (Δhmgb2 ANKA), an effect associated with a reduction of histological brain lesions and with lower expression levels of several proinflammatory genes. The incidence of ECM in pbhmgb2-deficient mice was restored by the administration of recombinant PbHMGB2. Protection from experimental cerebral malaria in Δhmgb2 ANKA-infected mice was associated with reduced sequestration in the brain of CD4+ and CD8+ T cells, including CD8+ granzyme B+ and CD8+ IFN-γ+ cells, and, to some extent, neutrophils. This was consistent with a reduced parasite sequestration in the brain, lungs, and spleen, though to a lesser extent than in wild-type P. berghei ANKA-infected mice. In summary, Plasmodium HMGB2 acts as an alarmin that contributes to the pathogenesis of cerebral malaria.
... Unlike the P. yoelii 17XNL, infection by P. yoelii 17XL induces experimental cerebral malaria that is associated with sequestration of infected red blood cells in the microvasculature of the mouse brain (Sarfo et al., 2005). In this respect, the virulent mouse strain resembles human P. falciparum infection more closely than Plasmodium berghei ANKA infection that shows little, if any, inflammation of the brain (Kaul et al., 1994). The P. yoelii 17XL-infected rodent model also has been used to investigate the role of chemokines, such as regulated on activation normal T cell expressed and secreted, CC chemokine receptor (CCR)1, CCR3, and CCR5, in the experimental cerebral malaria (Sarfo et al., 2005). ...
Article
Full-text available
Plasmodium yoelii 17XL was used to investigate the mechanism of Plasmodium falciparum-caused cerebral malaria, although its histological effect on other mouse organs is still unclear. Here, histological examination was performed on mice infected with P. yoelii 17XL; the effect of P. yoelii 17XL infection on anemia and body weight loss, as well as its lesions in the brain, liver, kidney, lung, and spleen, also was investigated. Plasmodium yoelii 17XL-infected red blood cells were sequestered in the microcirculation of the brain and in the kidney. Compared with the nonlethal P. yoelii 17XNL strain, infection by P. yoelii 17XL caused substantial pulmonary edema, severe anemia, and significant body weight loss. Although P. yoelii 17XNL and 17XL produced a similar focal necrosis in the mouse liver, infection of P. yoelii 17XL induced coalescing of red and white pulp. Mortality caused by P. yoelii 17XL may be due to cerebral malaria, as well as respiratory distress syndrome and severe anemia. Plasmodium yoelii 17XL-infected rodent malaria seems to be a useful model for investigating severe malaria caused by P. falciparum.
... 35. Chen D, Kaul DK, 1994. Rheological and hemodynamic characteristics of red cells of mouse, rat and human. ...
Article
The cytoadherence of infected red blood cells (IRBCs) to the vascular endothelium is the major cause of IRBC sequestration and vessel blockage in the cerebral form of human malaria. Among the rodent models of malaria, Plasmodium yoelii 17XL-infected mice show many similarities with the human cerebral malaria caused by P. falciparum. In both, the sequestration of IRBCs in the brain vessels is secondary to the cytoadherence of IRBCs to the vascular endothelium. Similar to P. falciparum infection in the human but in contrast to P. berghei ANKA infection in mice, P. yoelii 17XL results in little, if any, accumulation of monocytes in the brain. In vivo microcirculatory studies reported here were designed to further understand the hemodynamic aspects and mechanisms underlying cytoadherence of IRBCs in the P. yoelii model using the easily accessible cremaster muscle vasculature. The results show significant decreases in arteriovenous red blood cell velocities (Vrbc) and wall shear rates in the microcirculation of P. yoelii-infected mice, with a maximal decrease occurring in small-diameter postcapillary venules, the main sites of cytoadherence. This reflects contributions from IRBC cytoadherence as well as from increased rigidity of parasitized red blood cells. No cytoadherence is observed in arterioles of the infected mice despite decreased wall shear rates, indicating that endothelial receptors for cytoadherence are restricted to venules. Infusion of a monoclonal antibody (MAb) against the intercellular adhesion molecule-1 (ICAM-1) resulted in significant increases in both arteriolar and venular Vrbc and wall shear rates, accompanied by detachment of adhered IRBCs at some venular sites. The peripheral blood smears taken after the MAb infusion showed a distinct increase in the percentage of schizonts, again indicating detachment and/or prevention of cytoadherence. An MAb against the vascular cell adhesion molecule-1 (VCAM-1) as well as an irrelevant control antibody had no effect on these parameters. These results provide the first in vivo microcirculatory evidence indicating involvement of ICAM-1, but not of VCAM-1, in the sequestration of IRBCs in a rodent model of cerebral malaria.
... Mice of the intermediate cluster had significantly more parasites in the brain than susceptible ones. Sequestration of parasites in the brain is mainly associated with human cerebral malaria (CM) but has also been reported in mice [42,43]. As we do not have data on cytokine levels, lymphocyte counts or any ECM markers for the different mouse strain, death could be due to various reasons. ...
Article
Full-text available
The genetic background of a patient determines in part if a person develops a mild form of malaria and recovers, or develops a severe form and dies. We have used a mouse model to detect genes involved in the resistance or susceptibility to Plasmodium berghei malaria infection. To this end we first characterized 32 different mouse strains infected with P. berghei and identified survival as the best trait to discriminate between the strains. We found a locus on chromosome 6 by linking the survival phenotypes of the mouse strains to their genetic variations using genome wide analyses such as haplotype associated mapping and the efficient mixed-model for association. This new locus involved in malaria resistance contains only two genes and confirms the importance of Ppar-gamma in malaria infection.
... Although more extensively studied as a model of hyperparasitaemia and failure of parasite control (Couper et al. 2007Couper et al. , 2008), PyXL has been shown to sequester within the brain microvasculature and produce a cerebral syndrome comparable with human cerebral malaria (Yoeli and Hargreaves, 1974 ; Kaul et al. 1994) ; however, the hyper-parasitaemia associated with this infection (rapidly ascending peripheral parasitaemia that can reach 80–100 %) is not typical of human CM cases (Silamut et al. 1999) and this model is not widely used to study CM. In a few studies, P. berghei K173 has been found to induce CM-like signs ( Mitchell et al. 2005), but the dose-dependent onset of ECM in this model (inducing cerebral pathology after low dose but not high dose infection) (Mitchell et al. 2005) also limits its utility as a model of human CM : indeed P. berghei K173 is frequently used as a non- ECM-infection to compare with the most widely used model of ECM, P. berghei ANKA infection (Mitchell et al. 2005). ...
Article
Full-text available
Cerebral malaria is a life-threatening complication of malaria infection. The pathogenesis of cerebral malaria is poorly defined and progress in understanding the condition is severely hampered by the inability to study in detail, ante-mortem, the parasitological and immunological events within the brain that lead to the onset of clinical symptoms. Experimental murine models have been used to investigate the sequence of events that lead to cerebral malaria, but there is significant debate on the merits of these models and whether their study is relevant to human disease. Here we review the current understanding of the parasitological and immunological events leading to human and experimental cerebral malaria, and explain why we believe that studies with experimental models of CM are crucial to define the pathogenesis of the condition.
... It has been proposed that the Plasmodium yoelii model is better than the Plasmodium berghei model because P. yoelii-parasitized erythrocytes , like P. falciparum-parasitized erythrocytes, bind to brain microvasculature. However, Hearn et al. reported that P. berghei-parasitized erythrocytes adhere to brain microvasculature , indicating that both P. yoelii and P. berghei mimic P. falciparum in this regard (15, 19, 20). We selected the P. berghei model for this study because P. berghei-infected mice develop impaired consciousness, whereas P. yoelii-infected mice do not (10, 28, 38). ...
Article
Full-text available
The Plasmodium berghei-infected mouse model is a well-recognized model for human cerebral malaria. Mice infected withP. berghei exhibit (i) metabolic acidosis (pH < 7.3) associated with elevated plasma lactate concentrations, (ii) significant (P < 0.05) vascular leakage in their lungs, hearts, kidneys, and brains, (ii) significantly (P < 0.05) higher cell and serum glutamate concentrations, and (iv) significantly (P < 0.05) lower mean arterial blood pressures. Because these complications are similar to those of septic shock, the simplest interpretation of these findings is that the mice develop shock brought on by the P. berghei infection. To determine whether the immune system and specifically CD8+ T cells mediate the key features of shock during P. berghei malaria, we depleted CD8+T cells by monoclonal antibody (mAb) treatment and assessed the complications of malarial shock. P. berghei-infected mice depleted of CD8+ T cells by mAb treatment had significantly reduced vascular leakage in their hearts, brains, lungs, and kidneys compared with infected controls treated with rat immunoglobulin G. CD8-depleted mice were significantly (P < 0.05) protected from lactic acidosis, glutamate buildup, and diminished HCO3 −levels. Although the blood pressure decreased in anti-CD8 mAb-treated mice infected with P. berghei, the cardiac output, as assessed by echocardiography, was similar to that of uninfected control mice. Collectively, our results indicate that (i) pathogenesis similar to septic shock occurs during experimental P. bergheimalaria, (ii) respiratory distress with lactic acidosis occurs duringP. berghei malaria, and (iii) most components of circulatory shock are ameliorated by depletion of CD8+ T cells.
... Therefore, the analysis of the interaction of the parasitized erythrocytes with host endothelium is practically restricted to the examination of postmortem samples. Rodent malaria models are useful for the study of many aspects of the disease, but the pathology of murine plasmodium infections is different from that of human cerebral malaria, and sequestration of parasitized erythrocytes to the brain vasculature has not been observed in the models used so far, except for Plasmodium yoelii XL, where infection was not associated with inflammatory reactions in the brain (9,10). An alternative is to investigate the adhesion of isolated P. fakiparum-infected erythrocytes to cultured cell lines in vitro. ...
Article
Full-text available
Cerebral malaria is a fatal complication of infection by Plasmodium falciparum in man. The neurological symptoms that characterize this form of malarial disease are accompanied by the adhesion of infected erythrocytes to the vasculature of the brain. To study this phenomenon in vivo, an acute phase severe combined immunodeficiency (SCID) mouse model was developed in which sequestration of P. falciparum-infected human erythrocytes took place. During acute cerebral malaria in humans, the expression of intercellular adhesion molecule-1 (ICAM-1) is induced in vascular endothelium by inflammatory reactions. Acute phase ICAM-1 expression can also be obtained in SCID mice. The endothelium of the midbrain region was the most responsive to such inflammatory stimulus. It is noteworthy that the reticular formation in the midbrain controls the level of consciousness, and loss of consciousness is a symptom of cerebral malaria. We found that infected human erythrocytes were retained 24 times more than normal erythrocytes in ICAM-1-positive mouse brain. Sequestration to the brain was reduced by anti-ICAM-1 antibodies. These in vivo results were confirmed by the binding of P. falciparum-infected erythrocytes to the ICAM-1-positive endothelium in tissue sections of mouse brain. We conclude that the SCID mouse serves as a versatile in vivo model that allows the study of P. falciparum-infected erythrocyte adhesion as it occurs in human cerebral malaria. Upregulation of ICAM-1 expression in the region of the midbrain correlates with increased retention of malaria-infected erythrocytes and with the symptoms of cerebral malaria.
... In this CM model, the petechial hemorrhages that are characteristic of CM were attributed to sequestration of mononuclear white cells which interact with, and infiltrate across, the endothelium and postcapillary venules (Grau et al., 1986;Curfs et al., 1989;Eling and Sauerwein, 1995). However, the hallmark of CM in human malaria is parasite sequestration (MacPherson et al., 1985), a property which has also been described recently in a rodent model (Kaul et al., 1994). The chelators were assessed as therapeutic agents via two modes of administration: by multiple i.p. injections and by s.c. or i.p. insertion of a single dose of drug (or combination of drugs) which was encapsulated into biodegradable polymers. ...
Article
Full-text available
The hydrophilic desferrioxamine (DFO) and the lipophilic salicylaldehyde isonicotinoyl hydrazone (SIH) are iron chelators which inhibit in vitro proliferation of Plasmodium falciparum with similar potency (IC50 approximately 20 microM in 24- to 48-h tests). The in vivo assessment of these drugs was performed on Swiss mice infected with Plasmodium vinckei petteri with novel modes of drug administration and release. The drugs were delivered postpatently either by multiple i.p. injections or by a single i.p. or s.c. insertion of a drug-containing polymeric device which released most of the drug within 7 days at apparently first-order rates. A regimen of three daily i.p injections of 5 mg DFO for 3 consecutive days or a 70-mg dose of the drug given as an i.p. or s.c. polymer implant evoked similar delay and reduction in peak parasitemias and reduced mortality with no apparent signs of toxicity. Relatively faster, but otherwise similar results were obtained with the less hydrophilic SIH. In combination, the two drugs apparently potentiated each other. The polymeric devices were particularly useful for treating Plasmodium berghei K173-infected C57Bl mice, a suggested model of cerebral malaria, in which classical methods of DFO delivery were ineffective. The insertion of a 140-mg DFO-containing device on day 6 postinfection (parasitemia approximately 1%) led to a marked reduction in parasite proliferation, appearance of neurological sequelae and mortality of mice. Our studies indicate that polymeric devices for slow drug release might be highly advantageous for both hydrophilic and lipophilic drugs whose antimalarial efficacy might depend on the maintenance of sustained blood levels. The results obtained with slow-release devices have implications for malaria chemotherapy as well as for iron chelation therapy in iron overload conditions.
... Obviously then, cytoadherence with or without rosetting should increase resistance to blood flow through the venules. This was indeed shown to be the case under flow conditions in various experimental systems [36,[38][39][40][41][42]. Histology, however, indicates that even in severe malaria not all venules are obstructed. ...
Article
Full-text available
The development and propagation of malaria parasites in their vertebrate host is a complex process in which various host and parasite factors are involved. Sometimes the evolution of parasitaemia seems to be quelled by parasite load. In order to understand the typical dynamics of evolution of parasitaemia, various mathematical models have been developed. The basic premise ingrained in most models is that the availability of uninfected red blood cells (RBC) in which the parasite develops is a limiting factor in the propagation of the parasite population. We would like to propose that except in extreme cases of severe malaria, there is no limitation in the supply of uninfected RBC for the increase of parasite population. In this analysis we examine the biological attributes of the parasite-infected RBC such as cytoadherence and rosette formation, and the rheological properties of infected RBC, and evaluate their effects on blood flow and clogging of capillaries. We argue that there should be no restriction in the availability of uninfected RBC in patients. There is no justification for the insertion of RBC supply as a factor in mathematical models that describe the evolution of parasitaemia in the infected host. Indeed, more recent models, that have not inserted this factor, successfully describe the evolution of parasitaemia in the infected host.
Article
Plasmodium berghei-infected mice, a well-recognized model of experimental cerebral malaria (ECM), exhibit a systemic inflammatory response. Most investigators hypothesize that leukocytes bind to endothelial cells via intercellular adhesion molecule 1 (ICAM-1), which causes endothelial damage, increased microvascular permeability, and, ultimately, death. ICAM-1-deficient mice on an ECM-susceptible C57BL/6 background were significantly ( p = .04) protected from P. berghei mortality compared with ICAM-1 intact controls. ICAM-1 expression assessed by the dual radiolabeled monoclonal antibody technique was increased in the brain and lung in C57BL/6 mice on day 6 of P. berghei infection compared with uninfected controls (5.3-fold, p = .0003 for brain and 1.8-fold, p = .04 for lung). The increase in ICAM-1 expression coincided with significant ( p < .05) increases in microvascular permeability in the brain and lung. In contrast to the hypothesized role for ICAM-1, in vivo analysis by intravital microscopy of leukocyte rolling and adhesion in brain microvasculature of mice revealed markedly increased levels of leukocyte rolling and adhesion in ICAM-1-deficient mice on day 6 of P. berghei infection compared with uninfected controls. In addition, ICAM-1 expression and microvascular permeability were increased in infected ECM-resistant BALB/c mice compared with uninfected BALB/c controls. These results collectively indicate that although ICAM-1 contributes to the mortality of experimental malaria, it is not sufficient for the development of severe experimental malaria. In addition, ICAM-1 expressed on the endothelium or on leukocytes is not required for leukocyte rolling or adhesion to the brain microvasculature of mice during P. berghei malaria. Leukocyte rolling and adhesion in the brain vasculature during P. berghei malaria use different ligands than observed during inflammation in other vascular beds.
Chapter
Unique to Plasmodium falciparum malaria parasites, the mature asexual stages of the life cycle are absent from the peripheral blood stream. Using syringe pumps and commercially available microslides, it is possible to mimic the blood flow, and investigate the interactions of erythrocytes infected by well-defined P. falciparum isolates for their ability to bind to various tissue receptors under physiological flow conditions. This chapter outlines the techniques needed to investigate how parasites bind to endothelial cells under physiological sheer stress conditions.
Article
Usualmente, a malária se manifesta como uma tríade clássica de sintomas, podendo evoluir para formas graves, sendo a malária cerebral a mais letal das complicações da doença causada pelo Plasmodium falciparum. Pesquisas em modelos experimentais ajudam a compreender os diversos mecanismos de evolução, tratamentos, aspectos genéticos e moleculares e déficits cognitivo-comportamentais associados à malária cerebral. Nossa equipe vem desenvolvendo, no Laboratório de Pesquisa em Malária do Instituto Oswaldo Cruz, Fiocruz (LPM, IOC, Fiocruz), estudos sobre o efeito de estímulos imunes no comportamento animal, com foco no comprometimento da memória e na manifestação de ansiedade, relatados como associados mesmo a formas não graves de malária em humanos. A estratégia adotada, o uso de modelos experimentais murinos suscetíveis ao desenvolvimento de malária cerebral, os estudos em perspectiva pelo nosso grupo e outros aspectos relacionados são abordados nesse breve sobrevoo da literatura específica recente.
Article
Studies in vitro by Pasvol et al (Nature, 270:171, 1977) have indicated that the growth of Plasmodium falciparum in cells containing fetal hemoglobin (HbF = α2γ2) is retarded, but invasion is increased, at least in newborn cells. Normal neonates switch from about 80% HbF at birth to a few percent at the end of the first year of life. Carriers of β-thalassemia trait exhibit a delay in the normal HbF switch-off, which might partially explain the protection observed in populations with this gene. To study this hypothesis in vivo, we used transgenic (γ) mice expressing human Aγ and Gγ chains resulting in 40% to 60% α2Mγ2 hemoglobin, infected with rodent malaria. Two species of rodent malaria were studied.P chabaudi adami causes a nonlethal infection, mainly in mature red blood cells (RBC). P yoelii 17XNL is a nonlethal infection, invading primarily reticulocytes, whereas P yoelii 17XL is a lethal variant of P yoelii 17XNL and causes death of mice in approximately 1 to 2 weeks. Data indicate that this strain may cause a syndrome resembling cerebral malaria caused by P falciparum (Am J Trop Med Hyg, 50:512, 1994). In γ transgenic mice infected with P chabaudi adami, the parasitemia rose more quickly (in agreement with Pasvol) than in control mice, but was cleared more rapidly. In mice infected with P yoelii 17XNL, a clear reduction in parasitemia was observed. Interestingly, splenectomy before this infection, did not reverse protection. The most striking effect was in lethal P yoelii17XL infection. Control mice died between 11 to 13 days, whereas γ mice cleared the infection by day 22 and survived, a phenomenon also observed in splenectomized animals. These results suggest that HbF does indeed have a protective effect in vivo, which is not mediated by the spleen. In terms of mechanisms, light microscopy showed that intraerythrocytic parasites develop slowly in HbF erythrocytes, and electron microscopy showed that hemozoin formation was defective in transgenic mice. Finally, digestion studies of HbF by recombinant plasmepsin II demonstrated that HbF is digested only half as well as hemoglobin A (HbA). We conclude that HbF provides protection from P falciparum malaria by the retardation of parasite growth. The mechanism involves resistance to digestion by malarial hemoglobinases based on the data presented and with the well-known properties of HbF as a super stable tetramer. In addition, the resistance of normal neonates for malaria can now be explained by a double mechanism: increased malaria invasion rates, reported in neonatal RBC, will direct parasites to fetal cells, as well as F cells, and less to the ≈20% of HbA containing RBC, amplifying the antimalarial effects of HbF.
Chapter
About 1% out of the 120 million new P. Jalciparwn infections that occur globally each year have been estimated to be complicated by severe manifestations leading to death. This gives an annual incidence of about 1–2 million cases (WHO, 1992) corresponding to one death due to malaria every 15–30 seconds (Greenwood et al., 1987). Although an astonishing figure it is most likely an underestimation as under-reporting is common in the most affected areas where the primary health care is inadequate or non-existent.
Thesis
Full-text available
MBChB, M.Med (Paediatrics) U.O.N (Registration Number H80/8457/02)
Thesis
Full-text available
MBChB, M.Med (Paediatrics) U.O.N (Registration Number H80/8457/02)
Article
The challenge of correctly identifying malaria infection continues to impede our efforts to control this disease. Recent studies report highly specific retinal changes in severe malaria patients; these retinal changes may represent a very useful diagnostic indicator for this disease. To further explore the ocular manifestations of malaria, we used hyperspectral imaging to study retinal changes caused by Plasmodium berghei ANKA parasitization in a mouse model. We collected the spectral reflectance of the ocular fundus from hyperspectral images of the mouse eye. The blood oxygen sensitive spectral region was normalized for variances in illumination, and used to calculate relative values that correspond to oxygenated hemoglobin levels. Oxygen hemoglobin levels are markedly lower in parasitized mice, indicating that hemoglobin digestion by P. berghei may be detected using spectral reflectance. Furthermore, the ocular reflectance of parasitized mice was abnormally elevated between 660nm and 750nm, suggesting fluorescence in this region. While the source of this fluorescence is not yet clear, its presence correlates strongly with P. Berghei parasitization, and may indicate the presence of hemozoin deposits in the retinal vasculature. The pathology of severe malaria still presents many questions for clinicians and scientists, and our understanding of cerebral malaria has been generally confined to clinical observation and postmortem examination. As the retina represents a portion of the central nervous system that can be easily examined noninvasively, our technique may provide the basis for an automated tool to detect and examine severe malaria via retinal changes.
Article
Malaria, a disease caused by the Plasmodium parasite, remains one of the most deadly infectious diseases known to mankind. The parasite has a complex life cycle, of which only the erythrocytic stage of the parasite's life cycle is responsible for the diverse pathologies induced during infection. To date, the disease mechanisms that underlie these pathologies are still poorly understood. In the case of infections caused by Plasmodium falciparum, the species responsible for most malaria related deaths, pathogenesis is thought to be due to the sequestration of infected red blood cells (IRBC) in deep tissues. Other human and rodent malaria parasite species are also known to exhibit sequestration. Here, we review the different techniques that allow researchers to study how rodent malaria parasites modify their host cells, the distribution of IRBCs in vivo as well as the interactions between IRBCs and host tissues.
Article
Full-text available
Aims: Erythrocyte complement regulatory proteins, complement receptor 1 (CR1) and decay accelerating factor (CD55) protect red blood cells (RBCs) from complement mediated damage by controlling complement activation cascade and potentially protect RBCs from complement mediated damage that may occur when immune complexes are Research Article International Journal of TROPICAL DISEASE & Health, 3(2): 133-147, 2013 134 formed following malaria infection. Given the important role of RBCs in regulation of complement activation, we considered the competence of sickle cell trait RBCs in these functions. Methods: Children (age 0-192 months; n=116) were enrolled in a nested case controlled study conducted in Kombewa Division, Kisumu west District between October and December 2004. Based on hemoglobin (Hb) type, children were stratified into those with HbAS (n=47) and HbAA (n=69). The 47 HbAS individuals were matched to the 69 HbAA individuals of similar age (± 2 months or ± 24 months for those below or more than 192 months, respectively) at a ratio of 1:1 or 1:2. Circulating CR1 levels and CD levels were quantified using a FACScan cytometer under normal and reduced oxygen saturation. Results: The mean CR1 copy numbers per RBC was comparable in the two groups. However, between the ages of 49-192 months, the mean CR1 copy numbers per erythrocyte was significantly higher in children who had HbAS compared to those with HbAA (P=0.0332). The mean CD55 levels were comparable between the two groups but after deoxygenation, the mean CD levels in RBCs of individuals with HbAS was significantly higher than in the HbAA (P=0.011). Conclusion: The mean CR1 and CD55 copy numbers per RBC were comparable between the two groups under normal and reduced oxygen saturation. Beyond the age of 49 months, the CR1 copy numbers was higher in the HbAS compared to HbAA and this was also true for CD55 levels under deoxygenated conditions. Taken together, these results demonstrate that in the younger age groups, the protection afforded by HbAS against severe manifestations of malaria may be due to other factors other than complement regulatory proteins but beyond the age of 49 months, this protection may be partly due to the high CR1 copy numbers in the HbAS individuals.
Article
Full-text available
Plasmodium falciparum malaria is responsible for nearly one million annual deaths worldwide. Because of the difficulty in monitoring the pathogenesis of cerebral malaria in humans, we conducted a study in various mouse models to better understand disease progression in experimental cerebral malaria (ECM). We compared the effect on the integrity of the blood brain barrier (BBB) and the histopathology of the brain of P. berghei ANKA, a known ECM model, P. berghei NK65, generally thought not to induce ECM, P. yoelii 17XL, originally reported to induce human cerebral malaria-like histopathology, and P. yoelii YM. As expected, P. berghei ANKA infection caused neurological signs, cerebral hemorrhages, and BBB dysfunction in CBA/CaJ and Swiss Webster mice, while Balb/c and A/J mice were resistant. Surprisingly, PbNK induced ECM in CBA/CaJ mice, while all other mice were resistant. P. yoelii 17XL and P. yoelii YM caused lethal hyperparasitemia in all mouse strains; histopathological alterations, BBB dysfunction, or neurological signs were not observed. Intravital imaging revealed that infected erythrocytes containing mature parasites passed slowly through capillaries making intimate contact with the endothelium, but did not arrest. Except for relatively rare microhemorrhages, mice with ECM presented no obvious histopathological alterations that would explain the widespread disruption of the BBB. Intravital imaging did reveal, however, that postcapillary venules, but not capillaries or arterioles, from mice with ECM, but not hyperparasitemia, exhibit platelet marginalization, extravascular fibrin deposition, CD14 expression, and extensive vascular leakage. Blockage of LFA-1 mediated cellular interactions prevented leukocyte adhesion, vascular leakage, neurological signs, and death from ECM. The endothelial barrier-stabilizing mediators imatinib and FTY720 inhibited vascular leakage and neurological signs and prolonged survival to ECM. Thus, it appears that neurological signs and coma in ECM are due to regulated opening of paracellular-junctional and transcellular-vesicular fluid transport pathways at the neuroimmunological BBB.
Article
Cerebral malaria (CM) is a complex condition whose pathogenesis is still poorly understood. Our current knowledge is based on input from clinical, autopsy and genetic studies and from research in model systems, particularly in mice, which are well characterised in terms of histopathology, gene deficiencies and immune response pathways. This short review outlines several in vivo and in vitro models that are highly appropriate for interdisciplinary approaches for the investigation of CM pathogenesis. Section Editors:
Chapter
Full-text available
Malaria remains a major global health problem and cerebral malaria is one of themost serious complications of this disease. Recent years have seen important advances in our understanding of the pathogenesis of cerebralmalaria. Extensive analysis of tissues and blood taken frompatients with cerebralmalaria has been complimented by the use of animal models to identify specific components of pathogenic pathways. In particular, an important role for CD8+ T cells has been uncovered, as well divergent roles for members of the tumor necrosis factor (TNF) family of molecules, including TNF and lymphotoxin alpha. It has become apparent that theremay bemore than one pathogenic pathway leading to cerebral malaria. The last few years have also seen the testing of vaccines designed to target malaria molecules that stimulate inflammatory responses and thereby prevent the development of cerebral malaria. In this review, we will discuss the above advancements, as well as other important findings in research into the pathogenesis of cerebral malaria. As our understanding of pathogenic responses to Plasmodium parasites gathers momentum, the chance of a breakthrough in the development of treatments and vaccines to prevent death fromcerebralmalaria have become more realistic.
Article
Cerebral malaria is an acute encephalopathy evolving from an infection with Plasmodium falciparum which kills more than one million people each year. Brain tissues from patients who died with cerebral malaria revealed multifocal capillary obstruction by parasitised red blood cells, platelets, and leukocytes. Many studies are unified in their proposal of two major hypotheses consisting of cell adhesion to the brain endothelium and excessive immune stimulation resulting in further vascular inflammation, prothrombotic cell activation, mechanical obstruction of cerebral capillaries and, consequently, blood-brain barrier disruption. Platelets and endothelial cells communicate on multiple levels. Infection-induced changes in platelets and endothelial cells occur in cerebral malaria, resulting in their concomitant activation, increased interactions between these two cell types, and a secondary procoagulant or hypercoagulable state. Here we review evidence for these mechanisms and highlight the possible role of platelets as effectors of endothelial damage in cerebral malaria. A better understanding of the complex regulation of these various interactions between brain endothelial cells and platelets in the context of cerebral malaria may prove useful in the development of new approaches to the treatment of this disease.
Article
Full-text available
Previous studies from our laboratories have shown that transgenic mice expressing high levels of beta S globin are well-protected from Plasmodium chabaudi adami and partially protected against P berghei (Shear et al, Blood 81:222, 1993). We have now infected transgenic mice expressing low (39%), intermediate (57%), and high (75%) levels of beta S with the virulent strain of P yoelii (17XL) that appears to cause cerebral malaria. We find that the level of protection in these three groups of mice correlates positively with the level of beta S chain expression in the mice. Seven of nine mice expressing the high level of beta S recovered from infection, as did 7 of 9 mice expressing the intermediate level of beta S. Control mice and mice expressing the lower level of beta S all succumbed to infection. In mice expressing high and intermediate levels of beta S, parasites were found almost exclusively in reticulocytes during recovery, suggesting that mature red blood cells expressing beta S are more resistant than reticulocytes. These studies confirm epidemiologic data and offer insight into the mechanism of protection of sickle trait individuals against falciparum malaria.
Article
Insect-reared Anopheles gambiae were experimentally fed with the blood of naturally infected human volunteers carrying gametocytes of Plasmodium falciparum. Infection of at least one mosquito was successful in 86 experiments. For these gametocyte carriers, the hemoglobin types studied were AA (normal, n = 77), AS (heterozygous sickle cell, n = 8), and SS (homozygous sickle cell, n = 1). The mean of the percentages of infected mosquitoes by gametocyte carriers of AS hemoglobin was almost double that of carriers of AA: 30.4% versus 17.5%. The genetic protection in humans conferred by the beta(s) gene in its heterozygous form seems to be associated with an increasing effect on P. falciparum transmission from humans to mosquitoes. The epidemiologic and evolutionary aspects of this finding are discussed.
Article
Full-text available
Intravascular sickling, red cell-endothelium interaction, and altered microvascular responses have been suggested to contribute to the pathophysiology of human sickle cell disease, but have never been demonstrated under in vivo flow. To address this issue, we have examined a transgenic mouse line, alphaHbetaSbetaS-Antilles [betaMDD] which has a combined high (78%) expression of beta S and beta S-Antilles globins. In vivo microcirculatory studies using the cremaster muscle preparation showed adhesion of red cells, restricted to postcapillary venules, in transgenic mice but not in control mice. Electron microscopy revealed distinct contacts between the red cell membrane and the endothelium surface. Some red cells exhibiting sickling were regularly observed in the venular flow. Infusion of transgenic mouse red cells into the ex vivo mesocecum vasculature also showed adhesion of mouse red cells exclusively in venules. Under resting conditions (pO2, 15-20 mmHg), there were no differences in the cremaster microvascular diameters of control and transgenic mice; however, transgenic mice showed a drastic reduction in microvascular red cell velocities (Vrbc) with maximal Vrbc decrease (> 60%) occurring in venules, the sites of red cell adhesion and sickling. Local, transient hyperoxia (pO2, 150 mmHg) resulted in striking differences between control and transgenic mice. In controls, oxygen caused a 69% arteriolar constriction, accompanied by 75% reduction in Vrbc. In contrast, in transgenic mice, hyperoxia resulted in only 8% decrease in the arteriolar diameter and in 68% increase in VrBC; the latter is probably due to an improved flow behavior of red cells as a consequence of unsickling. In summary, the high expression of human sickle hemoglobin in the mouse results not only in intravascular sickling but also red cell-endothelium interaction. The altered microvascular response to oxygen could be secondary to blood rheological changes, although possible intrinsic differences in the endothelial cell/vascular smooth muscle function in the transgenic mouse may also contribute. These sickle transgenic mice could serve as a useful model to investigate vasoocclusive mechanisms, as well as to test potential therapies.
Article
Studies in vitro by Pasvol et al (Nature, 270:171, 1977) have indicated that the growth of Plasmodium falciparum in cells containing fetal hemoglobin (HbF = alpha2gamma2) is retarded, but invasion is increased, at least in newborn cells. Normal neonates switch from about 80% HbF at birth to a few percent at the end of the first year of life. Carriers of beta-thalassemia trait exhibit a delay in the normal HbF switch-off, which might partially explain the protection observed in populations with this gene. To study this hypothesis in vivo, we used transgenic (gamma) mice expressing human Agamma and Ggamma chains resulting in 40% to 60% alpha2Mgamma2 hemoglobin, infected with rodent malaria. Two species of rodent malaria were studied. P chabaudi adami causes a nonlethal infection, mainly in mature red blood cells (RBC). P yoelii 17XNL is a nonlethal infection, invading primarily reticulocytes, whereas P yoelii 17XL is a lethal variant of P yoelii 17XNL and causes death of mice in approximately 1 to 2 weeks. Data indicate that this strain may cause a syndrome resembling cerebral malaria caused by P falciparum (Am J Trop Med Hyg, 50:512, 1994). In gamma transgenic mice infected with P chabaudi adami, the parasitemia rose more quickly (in agreement with Pasvol) than in control mice, but was cleared more rapidly. In mice infected with P yoelii 17XNL, a clear reduction in parasitemia was observed. Interestingly, splenectomy before this infection, did not reverse protection. The most striking effect was in lethal P yoelii 17XL infection. Control mice died between 11 to 13 days, whereas gamma mice cleared the infection by day 22 and survived, a phenomenon also observed in splenectomized animals. These results suggest that HbF does indeed have a protective effect in vivo, which is not mediated by the spleen. In terms of mechanisms, light microscopy showed that intraerythrocytic parasites develop slowly in HbF erythrocytes, and electron microscopy showed that hemozoin formation was defective in transgenic mice. Finally, digestion studies of HbF by recombinant plasmepsin II demonstrated that HbF is digested only half as well as hemoglobin A (HbA). We conclude that HbF provides protection from P falciparum malaria by the retardation of parasite growth. The mechanism involves resistance to digestion by malarial hemoglobinases based on the data presented and with the well-known properties of HbF as a super stable tetramer. In addition, the resistance of normal neonates for malaria can now be explained by a double mechanism: increased malaria invasion rates, reported in neonatal RBC, will direct parasites to fetal cells, as well as F cells, and less to the approximately 20% of HbA containing RBC, amplifying the antimalarial effects of HbF.
Article
Previous studies demonstrated that Plasmodium yoelii 17XL, a lethal strain of rodent malaria, causes a syndrome in SW mice that resembles human cerebral malaria. The mouse brain pathology is characterized by cytoadherence of parasitized erythrocytes. Here, the possible mechanisms mediating cerebral malaria in this model were studied and the results were compared with a nonlethal strain of this parasite, P. yoelii 17XNL (nonlethal), which does not cause cerebral malaria. Immunostaining for intercellular adhesion molecule-1 (ICAM-1) revealed an increase in expression of this protein in the small venules and capillaries of the brains of infected mice that increased with time after infection. Staining was more pronounced during the lethal infection than the nonlethal infection. Some staining with monoclonal antibody to vascular cell adhesion molecule-1 was also observed, but it was quantitatively less than ICAM-1 staining and was limited to larger venules. During the lethal infection, levels of tumor necrosis factor-alpha (TNF-alpha) increased rapidly, peaking on day 4. In contrast, mice infected with nonlethal P. yoelii had a slower serum TNF-alpha response that peaked on day 10, prior to the maximum parasitemia. In addition, mice with a targeted disruption of the TNF-alpha gene (TNF-alpha-/- mice) were infected with the lethal and nonlethal strains of P. yoelii 17X. The TNF-alpha-/- mice infected with the nonlethal parasite had significantly higher levels of parasitemia than controls, whereas TNF-alpha-/- mice infected with the lethal strain had slightly higher levels of infected erythrocytes but were equally susceptible to death from this infection. Thus, TNF-alpha does not appear to be essential in mediating death. These results demonstrate that P. yoelii 17XL infection has features in common with human cerebral malaria and suggest that this model may be useful in testing strategies to alleviate this syndrome.
Article
Full-text available
Adherence of erythrocytes infected with Plasmodium falciparum to microvascular endothelial cells (sequestration) is considered to play an important role in parasite virulence and pathogenesis. However, the real importance of sequestration for infection and disease has never been fully assessed. The absence of an appropriate in vivo model for sequestration has been a major barrier. We have examined the rodent malaria parasite Plasmodium chabaudi chabaudi AS in mice as a potential model. Erythrocytes infected with this parasite adhere in vitro to purified CD36, a critical endothelium receptor for binding P. falciparum-infected erythrocytes. P. c. chabaudi-infected erythrocytes adhere in vitro to endothelial cells in a gamma interferon-dependent manner, suggesting the involvement of additional adhesion molecules in the binding process, as is also the case with P. falciparum-infected cells. Furthermore, plasma or sera from infected and hyperimmune mice, respectively, have the ability to block binding of infected erythrocytes to endothelial cells. In vivo, erythrocytes containing mature P. c. chabaudi parasites are sequestered from the peripheral circulation. Sequestration is organ specific, occurring primarily in the liver, although intimate contact between infected erythrocytes and endothelial cells is also observed in the spleen and brain. The results are discussed in the context of the use of this model to study (i) the relationship between endothelial cell activation and the level of sequestration and (ii) the primary function of sequestration in malaria infection.
Article
Full-text available
A murine model that closely resembles human cerebral malaria is presented, in which characteristic features of parasite sequestration and inflammation in the brain are clearly demonstrable. “Young” (BALB/c × C57BL/6)F1 mice infected with Plasmodium berghei (ANKA) developed typical neurological symptoms 7 to 8 days later and then died, although their parasitemias were below 20%. Older animals were less susceptible. Immunohistopathology and ultrastructure demonstrated that neurological symptoms were associated with sequestration of both parasitized erythrocytes and leukocytes and with clogging and rupture of vessels in both cerebral and cerebellar regions. Increases in tumor necrosis factor alpha and CD54 expression were also present. Similar phenomena were absent or substantially reduced in older infected but asymptomatic animals. These findings suggest that this murine model is suitable both for determining precise pathogenetic features of the cerebral form of the disease and for evaluating circumventive interventions.
Article
Full-text available
Malaria still is a major public health problem, partly because the pathogenesis of its major complication, cerebral malaria, remains incompletely understood. Experimental models represent useful tools to better understand the mechanisms of this syndrome. Here, data generated by several models are reviewed both in vivo and in vitro; we propose that some pathogenic mechanisms, drawn from data obtained from experiments in a mouse model, may be instrumental in humans. In particular, tumor necrosis factor (TNF) receptor 2 is involved in this syndrome, implying that the transmembrane form of TNF may be more important than the soluble form of the cytokine. It has also been shown that in addition to differences in immune responsiveness between genetically resistant and susceptible mice, there are marked differences at the level of the target cell of the lesion, namely, the brain endothelial cell. In murine cerebral malaria, a paradoxical role of platelets has been proposed. Indeed, platelets appear to be pathogenic rather than protective in inflammatory conditions because they can potentiate the deleterious effects of TNF. More recently, it has been shown that interactions among platelets, leukocytes, and endothelial cells have phenotypic and functional consequences for the endothelial cells. A better understanding of these complex interactions leading to vascular injury will help improve the outcome of cerebral malaria.
Article
Cerebral malaria is a serious and often fatal complication of Plasmodium falciparum infections. The precise mechanisms involved in the onset of neuropathology remain unknown, but parasite sequestration in the brain, metabolic disturbances and host immune responses are all thought to be involved. This review outlines the current state of knowledge of cerebral disease in humans, and discusses the contribution of studies of animal models to elucidation of the underlying mechanisms. © 2002 Éditions scientifiques et médicales Elsevier SAS. All rights reserved.
Article
Full-text available
Plasmodium berghei-infected mice, a well-recognized model of experimental cerebral malaria (ECM), exhibit many of the hallmarks of a systemic inflammatory response, with organ damage in brain, lung, and kidneys. Identification of the molecules mediating pathogenesis of the inflammatory response, such as leukocyte adhesion, may lead to new therapies. Indeed, mice lacking the cell adhesion molecule P-selectin were significantly (P = 0.005) protected from death due to P. berghei malaria compared with C57BL/6 controls despite similar parasitemia (P = 0.6) being found in both groups of mice. P-selectin levels assessed by the quantitative dual radiolabeled monoclonal antibody technique increased significantly (P < 0.05) in several organs in C57BL/6 mice infected with P. berghei, supporting the concept of a systemic inflammatory response mediating malarial pathogenesis. Intravital microscopic analysis of the brain microvasculature demonstrated significant (P < 0.001) leukocyte rolling and adhesion in brain venules of P. berghei-infected mice compared with those found in uninfected controls. The maximum leukocyte adhesion occurred on day 6 of P. berghei infection, when the mice become moribund and exhibit marked vascular leakage into the brain, lung, and heart. However, P-selectin levels were significantly (P < 0.005) increased in brain, lung, and kidneys during P. berghei malaria in ECM-resistant BALB/c mice compared with those found in uninfected BALB/c controls, indicating that increased P-selectin alone is not sufficient to mediate malarial pathogenesis. Leukocyte adhesion to brain microvessels of P-selectin-deficient mice with P. berghei malaria was similar to that observed in control mice. Collectively, these results indicate that P-selectin is important for the development of malarial pathogenesis but is not required for leukocyte adhesion in brain.
Article
A wide variety of transgenic mouse models expressing human globin genes have been generated in the last decade and a half. The majority of these models have been sickle transgenic models because they can advance our understanding of the pathophysiology of sickle cell disease, aid in development of therapeutic approaches, aid in development of preclinical strategies for antisickling gene therapy, and may be used for the identification and study of pleiotropic and epistatic genes.
Article
Plasmodium berghei-infected mice, a well-recognized model of experimental cerebral malaria (ECM), exhibit a systemic inflammatory response. Most investigators hypothesize that leukocytes bind to endothelial cells via intercellular adhesion molecule 1 (ICAM-1), which causes endothelial damage, increased microvascular permeability, and, ultimately, death. ICAM-1-deficient mice on an ECM-susceptible C57BL/6 background were significantly (p = .04) protected from P. berghei mortality compared with ICAM-1 intact controls. ICAM-1 expression assessed by the dual radiolabeled monoclonal antibody technique was increased in the brain and lung in C57BL/6 mice on day 6 of P. berghei infection compared with uninfected controls (5.3-fold, p = .0003 for brain and 1.8-fold, p = .04 for lung). The increase in ICAM-1 expression coincided with significant (p < .05) increases in microvascular permeability in the brain and lung. In contrast to the hypothesized role for ICAM-1, in vivo analysis by intravital microscopy of leukocyte rolling and adhesion in brain microvasculature of mice revealed markedly increased levels of leukocyte rolling and adhesion in ICAM-1-deficient mice on day 6 of P. berghei infection compared with uninfected controls. In addition, ICAM-1 expression and microvascular permeability were increased in infected ECM-resistant BALB/c mice compared with uninfected BALB/c controls. These results collectively indicate that although ICAM-1 contributes to the mortality of experimental malaria, it is not sufficient for the development of severe experimental malaria. In addition, ICAM-1 expressed on the endothelium or on leukocytes is not required for leukocyte rolling or adhesion to the brain microvasculature of mice during P. berghei malaria. Leukocyte rolling and adhesion in the brain vasculature during P. berghei malaria use different ligands than observed during inflammation in other vascular beds.
Article
Full-text available
Some patients with Plasmodium falciparum infections develop cerebral malaria, acute respiratory distress, and shock and ultimately die even though drug therapy has eliminated the parasite from the blood, suggesting that a systemic inflammatory response contributes to malarial pathogenesis. Plasmodium berghei-infected mice are a well-recognized model of severe malaria (experimental severe malaria [ESM]), and infected mice exhibit a systemic inflammatory response. Because platelets are proposed to contribute to ESM and other systemic inflammatory responses, we determined whether platelet adherence contributes to experimental malarial pathogenesis. Indeed, a significant (P < 0.005) increase in the number of rolling and adherent platelets was observed by intravital microscopy in brain venules of P. berghei-infected mice compared with the number in uninfected controls. P-selectin- or ICAM-1-deficient mice exhibit increased survival after P. berghei infection. We observed a significant (P < 0.0001) reduction in the morbidity of mice injected with anti-CD41 (αIIb or gpIIb) monoclonal antibody on day 1 of P. berghei infection compared with the morbidity of infected controls injected with rat immunoglobulin G. Additionally, platelet rolling and adhesion in brain venules were reduced in P. berghei mice lacking either P-selectin or ICAM-1 or when the platelets were coated with anti-CD41 monoclonal antibody. Unlike other inflammatory conditions, we did not detect platelet-leukocyte interactions during P. berghei malaria. Because (i) leukocyte adhesion is not markedly altered in the absence of P-selectin or ICAM-1 and (ii) CD41 is not an adhesion molecule for parasitized erythrocytes, these findings support the hypothesis that inhibition of platelet adhesion to the brain microvasculature protects against development of malarial pathogenesis.
Article
Sequestration of parasitized red blood cells in the cerebral vasculature is the predisposing event to the development of cerebral malaria during infection with Plasmodium falciparum. The adhesive interaction between these cells and receptors on the endothelial cell (cytoadhesion) occurs in the dynamic environment of the microcirculation, but most studies have neglected this factor and have concentrated on measuring adhesion in static (no flow) assays. Such studies ignore the markedly different rheological properties of parasitized red blood cells that become apparent when adhesion is examined under dynamic, flow conditions that resemble those of the circulation in vivo. Here, Brian Cooke and Ross Coppel review a number of novel aspects of cytoadhesion that have been identified using flow-based assays, and discuss their relevance to the pathophysiology, investigation and clinical management of falciparum malaria.
ResearchGate has not been able to resolve any references for this publication.