4,879
Views
98
CrossRef citations to date
0
Altmetric
Review

Cerebral malaria

Mysteries at the blood-brain barrier

, , , , , , & show all
Pages 193-201 | Published online: 01 Mar 2012

Abstract

Cerebral malaria is the most severe pathology caused by the malaria parasite, Plasmodium falciparum. The pathogenic mechanisms leading to cerebral malaria are still poorly defined as studies have been hampered by limited accessibility to human tissues. Nevertheless, histopathology of post-mortem human tissues and mouse models of cerebral malaria have indicated involvement of the blood-brain barrier in cerebral malaria. In contrast to viruses and bacteria, malaria parasites do not infiltrate and infect the brain parenchyma. Instead, rupture of the blood-brain barrier occurs and may lead to hemorrhages resulting in neurological alterations. Here, we review the most recent findings from human studies and mouse models on the interactions of malaria parasites and the blood-brain barrier, shedding light on the pathogenesis of cerebral malaria, which may provide directions for possible interventions.

Malaria remains one of the most prevalent infectious diseases in the world. The World Health Organization (WHO) reports that 50% of the world’s population living in 109 countries are still at risk of malaria.Citation1 More than 300 million clinical cases of malaria and one million deaths occur annually. Children under the age of 5 and pregnant women are most vulnerable to the disease. Hence, malaria continues to be a major global health problem, posing an enormous burden on mankind socially and economically.Citation2

The Malaria Parasite

Plasmodium, the infectious agent responsible for malaria, is transmitted by Anopheles spp mosquitoes. There are five species of Plasmodium spp infecting humans, with P. falciparum and P. vivax being the two most widespread.Citation1 The infection begins when an infected female Anopheles mosquito injects 5 to 50 sporozoites into the skin, which migrate to the liver.Citation3 Inside parenchymal hepatocyte, each sporozoite transforms to an exoerythrocytic parasite that multiplies giving rise to thousands of liver merozoites during the asymptomatic pre-erythrocytic phase. After maturation, liver merozoites are released into the blood stream where they infect red blood cells (RBC) and initiate the erythrocytic stage of the infection. The invading merozoite forms a vacuole, develops into a uninucleated ring form, then matures and divides into a multinucleated schizont. When the schizont ruptures, 4 to 16 merozoites are released into the bloodstream and infect new RBCs. During the blood stage, a subpopulation of merozoites will develop into gametocytes that will be taken up during a blood meal by mosquitoes, in which the sexual stage of the life cycle is completed.

The blood phase of the infection is responsible for the pathology of this disease. Symptoms of malaria usually develop 10–15 d after being bitten and include high fever, muscle aches and chills. In the majority of cases, infections are cleared by the use of appropriate treatments but in some patients, severe pathologies can develop and lead to death. Severe malaria includes a wide array of pathologies, ranging from metabolic alterations, renal failure, liver and lung dysfunctions, and anemia to cerebral malaria.Citation4

Human Cerebral Malaria

Human cerebral malaria (HCM) is the most severe complication of P. falciparum infection and has attracted the attention of both clinicians and scientists since the discovery of the malaria parasite.Citation4-Citation6 HCM can occur in less than two weeks after a mosquito bite and may develop after 2 to 7 d of fever.Citation4 The commonly accepted clinical definition of HCM is the neurological syndrome with patients in unrousable coma.Citation4,Citation7 Seizures, retinopathy and brainstem alterations due to elevated intracranial pressure and brain swelling are also clinical features frequently observed during HCM.Citation8,Citation9 To meet the HCM definition, the P. falciparum infection has to be confirmed and other causes of encephalitis (of viral or bacterial origins) to excluded. However, in field settings with limited resources, only easily diagnosed or obvious infectious diseases with brain involvement are investigated.Citation10-Citation12 Many viral, bacterial and parasitic infections can alter Plasmodium infections or pathologies and the reverse is also true.Citation12

Neurological symptoms are also frequently associated with severe metabolic acidosis, anemia and hypoglycemia.Citation13,Citation14 Thus exclusion of all these factors is important for the definition of “true HCM” and for making comparisons between different studies. It is well known that differences between pediatric and adult HCM exist.Citation15 Geographical differences in clinical patterns and prevalence of the syndrome are recognized and are likely due to differences in parasite and host genetics, immune status of the host, or epidemiological conditions.Citation4,Citation15 In many patients with HCM, death occurs rapidly before treatment can be administeredCitation16 and patients with HCM usually have a poor prognosis.Citation17 Patients who survive HCM may develop long-term neurological sequelaeCitation17,Citation18 and cognition and behavioral deficits.Citation19

There is no consensus on the pathogenesis of HCM. Indeed, this topic has been one of the most dogmatic and divisive in malaria research. This is due to the fact that limited studies can be performed in humans, while the common mouse model of cerebral malaria does not reproduce all aspects of HCM. The first attempt to uncover the pathogenesis of this syndrome has relied heavily on histopathology of brain tissues from patients who died of HCM. Since 1900, a series of studies with a limited number of patients have reported brain capillary occlusions, swelling of the endothelium, and sequestration mainly of infected red blood cells (IRBC).Citation20Citation25 In the past 20 years, studies with larger numbers of samples from patients with a more rigorous HCM definition have supported the original findings in most cases.Citation25Citation28 All these findings have led to the prevailing dogma that cerebral sequestration of IRBC is the etiologic mechanism leading to HCM.

The mechanisms by which sequestration leads to neurological complications and death are not yet clearly defined. It has been postulated that IRBC sequestration causes cerebral occlusion of brain capillaries, reduction of microvascular flow, decrease of nutrient supply to the brain, and damage to the vessel wall, leading to hemorrhages and neuronal alterations.Citation29 However, the most rigorous histology study performed so far on clinically confirmed HCM, showed no evidence of cerebral sequestration of P. falciparum in a proportion of the post-mortem brain tissue collected from Malawian children.Citation28 Still, it cannot be excluded that IRBC were sequestered before the patients were treated, and antimalarial treatment cleared the sequestered parasites but could not halt the cascade of events leading to HCM.

Leukocytes and platelets were found in the brain tissue of some of these Malawian children.Citation27,Citation30 Leukocytes have also been observed in the brains of adult patients from IndiaCitation31 but not from Thailand.Citation26,Citation32,Citation33 These different observations suggest that cerebral sequestration of IRBC may not always be uniquely responsible for HCM and that different or alternative pathological pathways leading to HCM may exist. They also demonstrate the limitations of making inferences based only on post-mortem histological studies, which cannot give any insight into the kinetics of the pathogenic processes occurring locally in the brain in order to identify the players involved. Other factors likely to be involved in HCM pathogenesis are proinflammatory and anti-inflammatory cytokines. However, their roles in HCM are still not clearly established. In some studies, HCM was associated with proinflammatory cytokines with HCM patients having higher levels of circulating TNF-α and/or IFN-γ,Citation34,Citation35 while in other studies, this was not observed.Citation36 One possible explanation for this discrepancy is the fact that in most reports, cytokine measurements were only performed at one time point at the time of diagnosis just before treatment. It is likely that some patients having high cytokine levels but not diagnosed as having HCM would have developed the syndrome if not treated. Another confounding factor is differences in HCM definition, in particular, the level of rigor in excluding co-infection (common in field studies) with pathogens that also modify the peripheral and local cytokine profiles. Nevertheless, the association of polymorphisms of pro-inflammatory cytokines or their receptorsCitation37 lends support to a role for these mediators.

Mouse Cerebral Malaria

A large body of research on cerebral malaria (CM) has been performed using mouse models of CM even though there have been strong controversies over the relevance of these models over the years.Citation38-Citation43 Although it is evident that mouse models do not replicate all aspects of the human disease, they have provided basic knowledge that can be applied to humans. Much of the debate on the value of mouse models is due to misconception and incomplete understanding concerning these models by clinicians or scientists working in HCM. However, on the other hand, a lack of rigorous characterization of these models and over-interpretation of data by researchers working on experimental cerebral malaria (ECM) further fueled the controversy. However, careful and balanced analyses of the data coupled with thorough understanding of parasite and mouse biology have allowed the validation of relevant hypotheses and the generation of new concepts applicable to HCM.

Of the four species of rodent malaria parasites, only a few P. berghei strains are able to induce ECM in mice with evident neurological involvement. The ANKA strain (PbA) has been the well-studied since its genome has been sequenced and analyzed extensively.Citation44 In addition, this is the strain of choice for genetic studies since transfection methods were first established for this strain.Citation45 ECM in PbA-infected susceptible mouse is characterized by the following neurological symptoms: paralysis, ataxia, deviation of the head and convulsion and/or coma.

In most susceptible mouse strains, 60 to 100 percent of the mice die of ECM during days 6 to 14 post-infection. The remaining mice die later at the end of week 2 or during the third week of infection due to hyperparasitemia and anemia.Citation46 ECM outcome can vary depending on the PbA parasite clone used,Citation47 the parasite form used for inoculation (sporozoite vs. IRBC),Citation48,Citation49 the dose of IRBC inoculatedCitation47 and the mode of propagation of the parasites, i.e., passage between different mouse strains.Citation47 PbA parasites, like their human Plasmodium counterparts, display phenotypic variationsCitation50,Citation51 and this influence the development of ECM.Citation47

Histopathological analyses of the brains of mice that developed ECM showed an accumulation of leukocytes and, to a lesser extent, of IRBC and normal RBC.Citation52-Citation54 Unlike in viral or bacterial infections, leukocytes do not infiltrate the parenchyma and are confined intravascularly. Normal RBC and IRBC are also located intravascularly unless hemorrhages have occurred.

Although accumulation of IRBC in the brains of PbA-infected mice is not as striking as in the brains of P. falciparum-infected patients who died of HCM, its occurrence has conclusively been demonstrated by quantitative PCRCitation53,Citation54 and dynamic bioluminescence imaging of transgenic PbA parasites expressing luciferase.Citation55CitationCitation57 PbA IRBC sequestration in mice is essential for ECM to occur since drug treatment just before neurological signs are expected prevents ECM.Citation57CitationCitation59 Sequestration is higher in susceptible C57BL/6J at the time of ECM signs than in resistant BALB/cJ mice (Claser and Renia, unpublished results) and is regulated by CD8+ T cells and IFN-γ.Citation55,Citation56 Very recent data have also shown that mature PbA IRBC cytoadhere to endothelial cells (Gruner, Ong and Renia, unpublished results), and mediate parasite sequestration. However, it has to be noted that only a fraction of PbA IRBC cytoadhere, explaining why mature blood forms of the parasite can be seen in the circulation unlike their P. falciparum counterparts.

Sequestered leukocytes are composed of monocytes and neutrophils (together representing 50 to 80% of the sequestered cells), CD4+ and CD8+ T cells, NK cells, platelets and few dendritic cells.Citation50,Citation60Citation63 Depletion of monocytes, neutrophils and platelets by antibody treatment a day or two before disease onset does not prevent the occurrence of ECM, suggesting that they have no effector functions during ECM.Citation46,Citation60,Citation63 However, they might have a role early in the infection through the production of cytokinesCitation63Citation66 or through their interaction with brain endothelial cells (see below). The role of NK cells is still unclear and debated. In one study, depletion of NK cells using anti-NK1.1 antibodies had no effect,Citation67 while in another study anti-asialo-GM1 antibodies prevented ECM by abrogating the migration of CD8+ T cells to the brain.Citation68 More work is needed using mice with specific deletion of NK cellsCitation69 to resolve this discrepancy since antibodies used for depletion can also deplete activated antigen-specific CD8+ T cells.Citation70

Depletion studies with specific antibodies and the use of mice deficient of immune cell subsets have shown that CD8+ T cells are the principal effector cells.Citation48,Citation60,Citation67,Citation71 Only 50,000 to 100,000 sequestered CD8+ T cells are found in a whole mouse brain at the time of ECM.Citation60 In addition, it is not known how many of these sequestered CD8+ T cells are parasite-specific, since PbA infection also induces non-specific activation of unrelated CD8+ T cells.Citation72 Depending on the parasite/mouse combination, CD4+ T cells can sometimes also behave as effector cells.Citation46,Citation63 However, in most combinations tested so far, CD4+ T cells play a role in the induction of ECMCitation60,Citation67,Citation73,Citation76 possibly by providing help for CD8+ T cells to fully mature.Citation74 Leukocytes are sequestered intravascularly and are in direct contact with endothelial cells. Thus, it has been postulated that CD8+ T cells might kill activated endothelial cells which have ingested and cross-presented cytoadherent parasites or parasite-derived material, thus disrupting the blood-brain barrier (BBB) and leading to neuronal dysfunction and death.Citation74,Citation75

The Blood-Brain Barrier and Malaria

The blood-brain barrier at homeostasis

The brain contains a network of blood vessels which are necessary for providing nutrients, and oxygen, and for removing carbon dioxide and waste (i.e., urea, creatinine, etc.). This network of capillaries together with the glia form a protective barrier called the blood-brain barrier (BBB). This barrier prevents large molecules and pathogens in the blood from entering the brain tissues and from altering the brain’s functions.Citation76,Citation77 The brain is very sensitive to blood chemistry variations and its homeostasis is tightly regulated.Citation78 The BBB is regarded as a part of the neurovascular unit, a concept that stresses a cross-talk between the different brain components for optimal functions of the brain. Maintenance of homeostasis is principally due to the brain endothelial cells, which are on the luminal side of the blood-brain barrier and correspond to the actual barrier site. Brain endothelial cells differ from those found in other tissues in many ways. They are attached by tight junctions of high electrical resistance preventing intercellular passage of molecules, and do not contain small openings called slit pores that allow the diffusion of molecules. Thus to reach the brain parenchyma, essential nutrients need to be actively transported by carrier systems to pass through the capillary wall. Brain endothelial cells also have important functions in mediating and regulating the immune response in the nervous system.Citation79 The inner part of the BBB is composed of pericytes, glial cells and astrocytes that essentially shield the capillaries from the neurons. The pericytes by themselves do not have a barrier function but contribute to the barrier function and phenotype of the endothelial cells. Astrocytes and glial cells contribute to homeostasis for neurological functioning by contributing to and regulating brain endothelial cell phenotype. In particular, endothelial cells are in contact with foot processes of astrocytes. These cellular structures provide an additional barrier protecting neurons from toxic products in the blood. Astrocytes can also form a barrier called the glia limitance at sites where the endothelial barrier is absent, such as the postrema.

The blood-brain barrier in HCM

A role for the BBB in HCM was postulated 40 years agoCitation80 and since then, many studies have been performed to uncover the extent of BBB alterations and their relationship to HCM pathogenic processes.Citation81-Citation83 Post-mortem observations on the accumulation of cytoadherent late-stage IRBC and of normal RBC in brain capillaries of infected humans have led to the prominent hypothesis that sequestration leads to capillary obstruction, reduced perfusion of the brain parenchyma and reduced delivery of necessary nutrients to neurons.Citation29 In addition, plugging of microvessels may also alter the function of BBB by creating local hypertension which increases pressure on tight junctions. If hypertension persists, tight junctions might break and this leads to the rupture of the BBB, causing hemorrhages. In addition, adhesion of IRBC to endothelial cells generates intracellular signaling in these cells, leading to activation and damage of the BBB. Although this hypothesis is attractive, it marginalizes the effects of systemic and local production of cytokines and parasite toxins such as malaria pigment, as well as the possible involvement of platelets and leukocytes that can also be found in the brain.Citation27,Citation30,Citation31

In an effort to determine if the BBB was altered in P. falciparum patients, measurements of molecules such albumin or immunoglobulins in the cerebrospinal fluid (CSF) and molecules excluded from the brain during homeostasis have been performed. In one study, radioactive-labeled albumin levels were not increased in the CSF after intravenous injection in Thai adult patients during and after coma.Citation84 In another study, albumin levels in the CSF of Vietnamese adult patients with HCM were not different from control subjects.Citation85 In Malawian children, the levels of albumin in the CSF were not different between children who died vs. those who survived, although they differ from UK adult controls.Citation89 When IgG was investigated, higher levels were detected in the CSF of a significant proportion in Thai patients with HCM,Citation87 but not in patients from Vietnam.Citation85 Discrepancies between these studies are probably due to measurements of samples obtained at a single time point. All together, these data suggest that although some BBB alterations occur during infection, major modifications leading to increased concentrations of plasma proteins in CSF as seen in bacterial or viral infections do not occur during HCM. However, focal ruptures of the BBB take place in the brain during HCM,Citation27 and may result from endothelial cell activation leading to the alteration of endothelial tight junctions, increased intercellular permeability, detachment from the basal matrix and/or death of endothelial cells. There is ample evidence that endothelial cells are activated during P. falciparum infection and during HCM as they show characteristic morphological modifications,Citation26 upregulate numerous surface antigens such as adhesion moleculesCitation32,Citation88 and produce a large variety of mediators.Citation89,Citation90

Activated endothelial cells in capillaries containing IRBC have also been shown to display a reduction but not a complete disappearance of cell-to-cell junction as shown by junction protein staining.Citation86,Citation91 In addition, perivascular macrophages in the vicinity of modified endothelial cells were also positive for macrophage activation markersCitation92 suggesting the passage of blood proteins into the perivascular space. This phenomenon seems to be restricted locally since it does not result in detectable leakage into the CNS. Nevertheless, the accumulation of localized increased permeability may lead to the activation of the microglia and/or astrocytes, which in turn may produce various mediators affecting vascular and neuronal cells. However, just an increase in permeability because of loosened tight junctions does not explain the hemorrhages observed in HCM histology studies. Hemorrhages result from focal rupture of the BBB and allow a large influx of plasma and plasma proteins to the interstitial brain tissue at these sites. This may explain the brain swelling, associated with coma, death or neurological sequela, which is frequently observed in patients with HCM.Citation93,Citation94 However, it has to be noted that brain swelling has been observed in the absence of hemorrhagesCitation8 and that coma is not caused by cerebral edema in Vietnamese patients who died of HCM.Citation95,Citation96

Numerous studies have been conducted to identify the mechanism involved in endothelial cell modifications. Proinflammatory plasma cytokines such as TNF-γ, Lymphotoxin, IFN-γ and IL-1β, which are increased during HCMCitation34,Citation35 can activate endothelial cells and modify vascular endothelium permeability.Citation96Citation99 There has been a lot of speculation regarding the role of nitric oxide (NO) in HCM.Citation100Citation104 The detection of inducible nitric oxide synthase in post-mortem brain tissues from African childrenCitation105 and Thai adultsCitation106 has suggested a pathogenic role for NO. However, although NO might participate in neuronal dysfunctions, it may have a protective role at the BBB level. It has been shown in vitro that NO can decrease permeability of the tight junctions induced by TNF-α or IFN-γ treatment.Citation107

Parasite cytoadherence can also directly activate endothelial cells as shown in vitro using brain endothelial cell lines.Citation108Citation114 This also leads to the production of chemokines and pro-inflammatory cytokines (IL-6, IL-8 and TNF-α), creating an activation loop.Citation110 Local productions of inflammatory cytokines and chemokines by endothelial cells and also microglial cells have been detected intravascularly in the brain of patients with HCM.Citation110,Citation114

P. falciparum IRBC express at their surface a variable parasite-derived antigen, P. falciparum erythrocyte membrane protein 1 (PfEMP1), encoded by the var multigene gene family.Citation115-Citation117 PfEMP-1 proteins have been shown to interact with a variety of surface receptors such as ICAM-1, thrombospondin, VCAM-1, PECAM-1, αvβ3 integrin, g1CR, endoglin, P-selectin, CD36 and fractalkine.Citation118-Citation121 Expression of some of these molecules is increased after cytokine stimulation.Citation79 When engaged, adhesion molecules like ICAM-1 can induce intracellular signaling which leads to the modification of cytoskeletal rearrangements.Citation122 However, co-engagement of CD31/PECAM can counteract ICAM-1 induced signaling.Citation123 During P. falciparum infection, clones expressing different PfEMP-1 variants with different adhesion receptor specificities can co-exist. A recent and elegant study has shown that upon interaction of IRBC with endothelial cells, materials can be transferred from the IRBC to the endothelial cell plasma membrane, which then endocytosed these materials. This is associated with opening of the intercellular tight junctions.Citation75 A more drastic effect of cytoadherence is induction of endothelial cell apoptosis.Citation124 This phenomenon has been described in vitro, is parasite strain-specific,Citation125 depends on host genetic factors,Citation126 and is mediated by a unique set of parasite proteinsCitation127 Different mechanisms leading to apoptosis have been described, involving the induction of oxidative stress and caspase-3.Citation102,Citation128

Platelet adhesion to brain endothelial cells can also lead to modifications of the BBB. They can facilitate IRBC adhesion by forming bridges between endothelial cells and IRBC, and they have been shown in vitro to potentiate endothelial cell apoptosis through TGF-β.Citation129 Expression of TGF-β has been detected in the brains of patients who died of HCM.Citation130 Leukocytes attracted by chemokines released at the site of IRBC adhesion can further induce modifications in endothelial cells by engaging ICAM-1 molecules and secreting proinflammatory cytokines and mediators locally.

An additional effect of sequestration of IRBC, platelets and leukocytes is a local reduction of blood flow.Citation131,Citation132 This creates a dysfunctional environment where toxins produced by metabolically-active parasites are concentrated and supply of oxygen and nutrients such as glucose and amino acids is decreased. A recent in vitro study has demonstrated that metabolic acidosis can increase endothelial intercellular permeability and disorganization of tight junctions.Citation113

The blood-brain barrier and ECM

Pioneer studies from Maegraith and collaborators first demonstrated that movement of both proteins and water did occur across the BBB during P. berghei infections using disulfine dye.Citation133 This phenomenon was further confirmed in later studies using Evans Blue, radio-labeled albumin, radio-labeled antibody or horseradish peroxidase, and was associated with brain edema in mice with ECM.Citation134-Citation136 Using retinal wholemounts, which allow the study of functional properties of microvasculature in a three-dimensional context, focal sites of BBB breakdown were observed in ECM-susceptible mice early in the infection.Citation137,Citation138 At later times of ECM, more extensive rupture of the BBB is seen and is associated with modifications of endothelial cell morphology such as swelling and signs of cell death, explaining the increase of protein and water transfer into the brain parenchyma.Citation134 This transfer led to an activation of astrocytes and microglia as shown in the retinal whole mount system.Citation139 In more recent studies, it was shown that during ECM, brain edema resulting from leakage and accumulation of fluid into the parenchymal extracellular space was observedCitation140,Citation141 and was associated with enlarged perivascular spaces.Citation142

Modifications of the BBB during ECM can be caused by various mechanisms. The early alterations of BBB may be caused by the activation of endothelial cells due to proinflammatory cytokinesCitation96,Citation143 produced in the first week of PbA infection.Citation64 TNF-α, lymphotoxin or IFN-γ affect endothelial cells by decreasing tight junction proteins while inducing an increase in the expression of adhesion moleculesCitation144 that mediate binding of leukocytes. Monocytes have been shown to adhere to endothelial cells in the retina as early as 2 d before disease onset, inducing morphology modifications of endothelial cells and provoking reduction of local blood flow.Citation137 Activated platelets also adhere to the brain microvasculature following TNF-α stimulation.Citation145,Citation146 They can also fuse with endothelial cells to increase leukocyte adhesion.Citation145 Of interest, an active role for NO has been discarded since ECM is characterized by low NO bioavailability like in HCM. And moreover, treatments with exogenous NO-donors prevent BBB rupture and protect against ECM.Citation147,Citation148

Although these early and focal modifications are important, they are not responsible for late ECM deaths. CD8+ T cells which migrate at the time of neurological signs are responsible for the ensuing lethality. It is not yet known if and how CD8+ T cells induce the rupture of the BBB, but it has been proposed that CD8+ T cells kill endothelial cells after they phagocytosed and processed parasite-derived antigens.Citation74 This hypothesis is supported by the fact that mice deficient for perforin or granzyme B, two CD8 cytotoxic effector molecules, are resistant to ECM.Citation58,Citation71,Citation149 However, a recent study in a mouse model of acute hemorrhagic leukoencephalomyelitis showed that CD8+ T cells altered BBB tight junction proteins through a perforin-dependent mechanism without inducing endothelial apoptosis.Citation150 It remains to be determined if such a mechanism exists in ECM.

Relevance of the ECM findings to HCM

The mouse model of malaria does not reproduce all the features of P. falciparum infection predominantly due to differences in parasite biology. However, similarities need to be recognized since they help to generate hypotheses that can be tested in humans. In summary, several pathologic changes occur mainly intravascularly in both ECM and HCM. The BBB is altered during infection and changes in permeability and/or rupture appear to be restricted locally, with BBB rupture leading to hemorrhages. The exact causes of BBB alterations are still unknown and may involve cytoadherent parasites, cytokines, platelets and/or leukocytes. The role of CD8+ T cells, which have been clearly demonstrated in the mouse model, is still strongly debated in HCM. The main refutation for a role of these cells is based on their rarity in post-mortem histology samples that represent a limited snapshot of the brain in both space and time. However, taking into account the low frequency of brain-sequestered mouse CD8+ T cells in ECM, human studies performed thus far may not have been sufficiently sensitive. Quantitative and carefully controlled immunohistological studies are needed, or alternatively, new molecular approaches such as quantification of CD8 mRNA in the brainCitation63 should be performed.

Conclusion

The BBB is an important protective barrier for the human host during malaria infections. Although the intra-erythrocytic parasites do not penetrate the brain parenchyma outside of local ruptures, there is a constant and dynamic interplay between IRBC, parasite-derived materials, host leukocytes and the BBB that can lead to neurological alterations and death. However, till now, it has not been firmly established if alterations of the BBB during HCM are key pathogenic factors. As such, there is a need to describe and understand the interactions of IRBC with the brain endothelium and their downstream effects on the microglia, astrocytes, and neurons and how they influence the morbidity and mortality during HCM. Comprehension of these pathways may pave the way for the development of new adjuvant therapies.

Acknowledgments

This work was supported by core funding from the Agency for Science, Technology and Research (A*STAR, Singapore).

References

  • Greenwood BM, Fidock DA, Kyle DE, Kappe SH, Alonso PL, Collins FH, et al. Malaria: progress, perils, and prospects for eradication. J Clin Invest 2008; 118:1266 - 76; http://dx.doi.org/10.1172/JCI33996; PMID: 18382739
  • Snow RW, Omumbo JA. Malaria. In: Jamison DT, Feachem RG, Makgoba MW, Bos ER, Baingana FK, Hofman KJ, Rogo KO, eds. Disease and Mortality in Sub-Saharan Africa. 2nd. Washington DC: World Bank; 2006.
  • Amino R, Menard R, Frischknecht F. In vivo imaging of malaria parasites - recent advances and future directions. Curr Opin Microbiol 2005; 8:407 - 14; http://dx.doi.org/10.1016/j.mib.2005.06.019; PMID: 16019254
  • Severe falciparum malaria. World Health Organization, Communicable Diseases Cluster. Trans R Soc Trop Med Hyg 2000; 94:S1 - 90; PMID: 11103309
  • Laveran A. Paludisme. In: Traite de Medecine et Therapeutique. 1 ed. Paris: BaillSres, J.-B. et Fils; 1897. p. 38-1154.
  • Marchiafava E, Bignami A. Malaria. Twentieth century practice of Medicine.London: Sampson Lowe; 1900.
  • Newton CRJ, Pasvol G, Winstanley PA, Warrell DA. Cerebral malaria: what is unarousable coma?. Lancet 1990; 335:472; http://dx.doi.org/10.1016/0140-6736(90)90703-8; PMID: 1968190
  • Idro R, Jenkins NE, Newton CRJ. Pathogenesis, clinical features, and neurological outcome of cerebral malaria. Lancet Neurol 2005; 4:827 - 40; http://dx.doi.org/10.1016/S1474-4422(05)70247-7; PMID: 16297841
  • Lewallen S, Bakker H, Taylor TE, Wills BA, Courtright P, Molyneux ME. Retinal findings predictive of outcome in cerebral malaria. Trans R Soc Trop Med Hyg 1996; 90:144 - 6; http://dx.doi.org/10.1016/S0035-9203(96)90116-9; PMID: 8761574
  • Berkley JA, Mwangi I, Mellington F, Mwarumba S, Marsh K. Cerebral malaria versus bacterial meningitis in children with impaired consciousness. QJM 1999; 92:151 - 7; http://dx.doi.org/10.1093/qjmed/92.3.151; PMID: 10326074
  • Marsh K, Forster D, Waruiru CM, Mwangi I, Winstanley MT, Marsh V, et al. Indicators of life-threatening malaria in african children. N Engl J Med 1995; 332:1399 - 404; http://dx.doi.org/10.1056/NEJM199505253322102; PMID: 7723795
  • Troye-Blomberg M, Berzins K. Immune interactions in malaria co-infections with other endemic infectious diseases: implications for the development of improved disease interventions. Microbes Infect 2008; 10:948 - 52; http://dx.doi.org/10.1016/j.micinf.2008.07.014; PMID: 18672089
  • Taylor TE, Wirima JJ, Molyneun ME, Davis TME, Brewster DR, Hill AVS, et al. Hypoglycaemia and cerebral malaria. Lancet 1990; 336:950 - 1; http://dx.doi.org/10.1016/0140-6736(90)92329-G; PMID: 1976969
  • Taylor TE, Borgstein A, Molyneux ME. Acid-base status in paediatric Plasmodium falciparum malaria. Q J Med 1993; 86:99 - 109; PMID: 8464997
  • Haldar K, Murphy SC, Milner DA, Taylor TE. Malaria: Mechanisms of Erythrocytic Infection and Pathological Correlates of Severe Disease. Annu Rev Pathol 2007; 2:217 - 49; http://dx.doi.org/10.1146/annurev.pathol.2.010506.091913; PMID: 18039099
  • Newton CRJ, Krishna S. Severe falciparum malaria in children: current understanding of pathophysiology and supportive treatment. Pharmacol Ther 1998; 79:1 - 53; http://dx.doi.org/10.1016/S0163-7258(98)00008-4; PMID: 9719344
  • Brewster DR, Kwiatkowski DP, White NJ. Neurological Sequelae of Cerebral Malaria in Children. Lancet 1990; 336:1039 - 43; http://dx.doi.org/10.1016/0140-6736(90)92498-7; PMID: 1977027
  • Idro R, Carter JA, Fegan G, Neville BG, Newton CRJ. Risk factors for persisting neurological and cognitive impairments following cerebral malaria. Arch Dis Child 2006; 91:142 - 8; http://dx.doi.org/10.1136/adc.2005.077784; PMID: 16326798
  • Holding PA, Snow RW. Impact of Plasmodium falciparum malaria on performance and learning: review of the evidence. Am J Trop Med Hyg 2001; 64:68 - 75; PMID: 11425179
  • Durk H. Uber die bei malaria comatosa aufretenden veranderungen des zentranervensystem. Archiv Schiffs Tropenhygien 1917; 21:117 - 32
  • Gaskell SJ, Millar WL. Studies on malignant malaaria in Macedonia. QJM 1920; 24:317 - 22
  • Kean BH, Smith JA. Death due to estivo-autumnal malaria. A resume of one hundred autopsy cases, 1925-1942. Am J Trop Med 1944; 24:317 - 22
  • Margulis MS. Zur frage der pathologish-anatomischen verunderungen bei busartige malaria. Neurologische Zentralblat 1914; 33:1019 - 24
  • Rigdon RH, Fletcher DE. Lesions of brain associated with malaria. Pathologic study on man and on experimental animals. Arch Neurol Psychiatry 1944; 53:191 - 8
  • Spitz S. Pathology of tropical diseases. Philadelphia: Saunders Co; 1961.
  • MacPherson GG, Warrell MJ, White NJ, Looareesuwan S, Warrell DA. Human cerebral malaria. A quantitative ultrastructural analysis of parasitized erythrocyte sequestration. Am J Pathol 1985; 119:385 - 401; PMID: 3893148
  • Dorovini-Zis K, Schmidt K, Huynh H, Fu W, Whitten RO, Milner D, et al. The neuropathology of fatal cerebral malaria in malawian children. Am J Pathol 2011; 178:2146 - 58; http://dx.doi.org/10.1016/j.ajpath.2011.01.016; PMID: 21514429
  • Taylor TE, Fu WJ, Carr RA, Whitten RO, Mueller JG, Fosiko NG, et al. Differentiating the pathologies of cerebral malaria by postmortem parasite counts. Nat Med 2004; 10:143 - 5; http://dx.doi.org/10.1038/nm986; PMID: 14745442
  • Berendt AR, Turner GDH, Newbold CI. Cerebral malaria: the sequestration hypothesis. Parasitol Today 1994; 10:412 - 4; http://dx.doi.org/10.1016/0169-4758(94)90238-0; PMID: 15275553
  • Grau GE, Mackenzie CD, Carr RA, Redard M, Pizzolato G, Allasia C, et al. Platelet accumulation in brain microvessels in fatal pediatric cerebral malaria. J Infect Dis 2003; 187:461 - 6; http://dx.doi.org/10.1086/367960; PMID: 12552430
  • Patnaik JK, Das BS, Mishra SK, Mohanty S, Satpathy SK, Mohanty D. Vascular clogging, mononuclear cell margination, and enhanced vascular permeability in the pathogenesis of human cerebral malaria. Am J Trop Med Hyg 1994; 51:642 - 7; PMID: 7985757
  • Silamut K, Phu NH, Whitty C, Turner GDH, Louwrier K, Mai NT, et al. A quantitative analysis of the microvascular sequestration of malaria parasites in the human brain. Am J Pathol 1999; 155:395 - 410; http://dx.doi.org/10.1016/S0002-9440(10)65136-X; PMID: 10433933
  • Pongponratn E, Turner GDH, Day NP, Phu NH, Simpson JA, Stepniewska K, et al. An ultrastructural study of the brain in fatal Plasmodium falciparum malaria. Am J Trop Med Hyg 2003; 69:345 - 59; PMID: 14640492
  • Grau GE, Taylor TE, Molyneux ME, Wirima JJ, Vassalli P, Hommel M, et al. Tumor necrosis factor and disease severity in children with falciparum Malaria. N Engl J Med 1989; 320:1586 - 91; http://dx.doi.org/10.1056/NEJM198906153202404; PMID: 2657427
  • Kwiatkowski D, Hill AVS, Sambou I, Twumasi PM, Castracane J, Manogue K, et al. TNF concentration in fatal cerebral, non fatal cerebral, and uncomplicated Plasmodium falciparum malaria. Lancet 1990; 336:1201 - 4; http://dx.doi.org/10.1016/0140-6736(90)92827-5; PMID: 1978068
  • Lyke KE, Burges R, Cissoko Y, Sangare L, Dao M, Diarra I, et al. Serum Levels of the Proinflammatory Cytokines Interleukin-1 Beta (IL-1beta), IL-6, IL-8, IL-10, Tumor Necrosis Factor Alpha, and IL-12(p70) in Malian Children with Severe Plasmodium falciparum Malaria and Matched Uncomplicated Malaria or Healthy Controls. Infect Immun 2004; 72:5630 - 7; http://dx.doi.org/10.1128/IAI.72.10.5630-5637.2004; PMID: 15385460
  • Campino S, Kwiatkowski DP, Dessein A. Mendelian and complex genetics of susceptibility and resistance to parasitic infections. Semin Immunol 2006; 18:411 - 22; http://dx.doi.org/10.1016/j.smim.2006.07.011; PMID: 17023176
  • Carvalho LJ. Murine cerebral malaria: how far from human cerebral malaria?. Trends Parasitol 2010; 26:271 - 2; http://dx.doi.org/10.1016/j.pt.2010.03.001; PMID: 20335068
  • Langhorne J, Buffet P, Galinski MR, Good MF, Harty J, Leroy D, et al. The relevance of non-human primate and rodent malaria models for humans. Malar J 2011; 10:23; http://dx.doi.org/10.1186/1475-2875-10-23; PMID: 21288352
  • Rénia L, Gruner AC, Snounou G. Cerebral malaria: in praise of epistemes. Trends Parasitol 2010; 26:275 - 7; http://dx.doi.org/10.1016/j.pt.2010.03.005; PMID: 20363672
  • Riley EM, Couper KN, Helmby H, Hafalla JC, de Souza JB, Langhorne J, et al. Neuropathogenesis of human and murine malaria. Trends Parasitol 2010; 26:277 - 8; http://dx.doi.org/10.1016/j.pt.2010.03.002; PMID: 20338809
  • Stevenson MM, Gros P, Olivier M, Fortin A, Serghides L. Cerebral malaria: human versus mouse studies. Trends Parasitol 2010; 26:274 - 5; http://dx.doi.org/10.1016/j.pt.2010.03.008; PMID: 20382077
  • White NJ, Turner GDH, Medana IM, Dondorp AM, Day NP. The murine cerebral malaria phenomenon. Trends Parasitol 2010; 26:11 - 5; http://dx.doi.org/10.1016/j.pt.2009.10.007; PMID: 19932638
  • Hall N, Karras M, Raine JD, Carlton JMR, Kooij TW, Berriman M, et al. A comprehensive survey of the Plasmodium life cycle by genomic, transcriptomic, and proteomic analyses. Science 2005; 307:82 - 6; http://dx.doi.org/10.1126/science.1103717; PMID: 15637271
  • Tomas AM, van der Wel AM, Thomas AW, Janse CJ, Waters AP. Transfection systems for animal models of malaria. Parasitol Today 1998; 14:245 - 9; http://dx.doi.org/10.1016/S0169-4758(98)01248-4; PMID: 17040769
  • Engwerda C, Belnoue E, Gruner AC, Renia L. Experimental models of cerebral malaria. Curr Top Microbiol Immunol 2005; 297:103 - 43; http://dx.doi.org/10.1007/3-540-29967-X_4; PMID: 16265904
  • Amani V, Boubou MI, Pied S, Marussig M, Walliker D, Mazier D, et al. Cloned lines of Plasmodium berghei ANKA differ in their abilities to induce experimental cerebral malaria. Infect Immun 1998; 66:4093 - 9; PMID: 9712753
  • Bagot S, Nogueira F, Collette A, Do Rosario VE, Lemonier F, Cazenave PA, et al. Comparative Study of Brain CD8+ T Cells Induced by Sporozoites and Those Induced by Blood-Stage Plasmodium berghei ANKA Involved in the Development of Cerebral Malaria. Infect Immun 2004; 72:2817 - 26; http://dx.doi.org/10.1128/IAI.72.5.2817-2826.2004; PMID: 15102792
  • Kordes M, Matuschewski K, Hafalla JC. Caspase-1 Activation of Interleukin-1beta (IL-1beta) and IL-18 Is Dispensable for Induction of Experimental Cerebral Malaria. Infect Immun 2011; 79:3633 - 41; http://dx.doi.org/10.1128/IAI.05459-11; PMID: 21708993
  • del Portillo HA, Fernandez-Becerra C, Bowman S, Oliver K, Preuss M, Sanchez CP, et al. A superfamily of variant genes encoded in the subtelomeric region of Plasmodium vivax. Nature 2001; 410:839 - 42; http://dx.doi.org/10.1038/35071118; PMID: 11298455
  • Roberts DJ, Craig AG, Berendt AR, Pinches RA, Nash GB, Marsh K, et al. Rapid switching to multiple antigenic and adhesive phenotypes in malaria. Nature 1992; 357:689 - 92; http://dx.doi.org/10.1038/357689a0; PMID: 1614515
  • Hearn J, Rayment N, Landon DN, Katz DR, de Souza JB. Immunopathology of Cerebral Malaria: Morphological Evidence of Parasite Sequestration in Murine Brain Microvasculature. Infect Immun 2000; 68:5364 - 76; http://dx.doi.org/10.1128/IAI.68.9.5364-5376.2000; PMID: 10948166
  • Jennings VM, Actor JK, Lal AA, Hunter RL. Cytokine profile suggesting that murine cerebral malaria is an encephalitis. Infect Immun 1997; 65:4883 - 7; PMID: 9353082
  • Hulier E, Petour P, Snounou G, Nivez MP, Miltgen F, Mazier D, et al. A method for the quantitative assessment of malaria parasite development in organs of the mammalian host. Mol Biochem Parasitol 1996; 77:127 - 35; http://dx.doi.org/10.1016/0166-6851(96)02584-4; PMID: 8813659
  • Amante FH, Haque A, Stanley AC, de Labastida Rivera F, Randall LM, Wilson YA, et al. Immune-Mediated Mechanisms of Parasite Tissue Sequestration during Experimental Cerebral Malaria. J Immunol 2010; 185:3632 - 42; http://dx.doi.org/10.4049/jimmunol.1000944; PMID: 20720206
  • Claser C, Malleret B, Gun SY, Wong AY, Chang ZW, Teo P, et al. CD8 T Cells and IFN-gamma Mediate the Time-Dependent Accumulation of Infected Red Blood Cells in Deep Organs during Experimental Cerebral Malaria. PLoS ONE 2011; 6:e18720; http://dx.doi.org/10.1371/journal.pone.0018720; PMID: 21494565
  • Baptista FG, Pamplona A, Pena AC, Mota MM, Pied S, Vigario AM. Accumulation of Plasmodium-infected red blood cells in the brain is crucial for the development of cerebral malaria in mice. Infect Immun 2010; 78:4033 - 9; http://dx.doi.org/10.1128/IAI.00079-10; PMID: 20605973
  • Haque A, Best SE, Unosson K, Amante FH, de Labastida F, Anstey NM, et al. Granzyme B Expression by CD8+ T Cells Is Required for the Development of Experimental Cerebral Malaria. J Immunol 2011; 186:6148 - 56; http://dx.doi.org/10.4049/jimmunol.1003955; PMID: 21525386
  • McQuillan JA, Mitchell AJ, Ho YF, Combes V, Ball HJ, Golenser J, et al. Coincident parasite and CD8+ T cell sequestration is required for development of experimental cerebral malaria. Int J Parasitol 2011; 41:155 - 63; http://dx.doi.org/10.1016/j.ijpara.2010.08.003; PMID: 20828575
  • Belnoue E, Kayibanda M, Vigario AM, Deschemin JC, van RN, Viguier M, et al. On the pathogenic role of brain-sequestered alphabeta CD8+ T cells in experimental cerebral malaria. J Immunol 2002; 169:6369 - 75; PMID: 12444144
  • Belnoue E, Kayibanda M, Deschemin JC, Viguier M, Mack M, Kuziel WA, et al. CCR5 deficiency decreases susceptibility to experimental cerebral malaria. Blood 2003; 101:4253 - 9; http://dx.doi.org/10.1182/blood-2002-05-1493; PMID: 12560237
  • Belnoue E, Costa FTM, Vigario AM, Voza T, Gonnet F, Landau I, et al. Chemokine receptor CCR2 is not essential for the development of experimental cerebral malaria. Infect Immun 2003; 71:3648 - 51; http://dx.doi.org/10.1128/IAI.71.6.3648-3651.2003; PMID: 12761155
  • Belnoue E, Potter SM, Rosa DS, Mauduit M, Gruner AC, Kayibanda M, et al. Control of pathogenic CD8+ T cell migration to the brain by IFN-gamma during experimental cerebral malaria. Parasite Immunol 2008; 30:544 - 53; http://dx.doi.org/10.1111/j.1365-3024.2008.01053.x; PMID: 18665903
  • van der Heyde HC, Gramaglia I, Sun G, Woods C. Platelet depletion by anti-CD41 (alphaIIb) mAb injection early but not late in the course of disease protects against Plasmodium berghei pathogenesis by altering the levels of pathogenic cytokines. Blood 2005; 105:1956 - 63; http://dx.doi.org/10.1182/blood-2004-06-2206; PMID: 15494426
  • Chen L, Zhang Z, Sendo F. Neutrophils play a critical role in the pathogenesis of experimental cerebral malaria. Clin Exp Immunol 2000; 120:125 - 33; http://dx.doi.org/10.1046/j.1365-2249.2000.01196.x; PMID: 10759773
  • Chen L, Sendo F. Cytokine and chemokine mRNA expression in neutrophils from CBA/NSlc mice infected with Plasmodium berghei ANKA that induces experimental cerebral malaria. Parasitol Int 2001; 50:139 - 43; http://dx.doi.org/10.1016/S1383-5769(01)00063-0; PMID: 11438437
  • Yañez DM, Manning DD, Cooley AJ, Weidanz WP, van der Heyde HC. Participation of lymphocyte subpopulations in the pathogenesis of experimental murine cerebral malaria. J Immunol 1996; 157:1620 - 4; PMID: 8759747
  • Hansen DS, Bernard NJ, Nie CQ, Schofield L. NK cells stimulate recruitment of CXCR3+ T cells to the brain during Plasmodium berghei-mediated cerebral malaria. J Immunol 2007; 178:5779 - 88; PMID: 17442962
  • Walzer T, Blery M, Chaix J, Fuseri N, Chasson L, Robbins SH, et al. Identification, activation, and selective in vivo ablation of mouse NK cells via NKp46. Proc Natl Acad Sci USA 2007; 104:3384 - 9; http://dx.doi.org/10.1073/pnas.0609692104; PMID: 17360655
  • Assarsson E, Kambayashi T, Sandberg JK, Hong S, Taniguchi M, Van Kaer L, et al. CD8+ T cells rapidly acquire NK1.1 and NK cell-associated molecules upon stimulation in vitro and in vivo. J Immunol 2000; 165:3673 - 9; PMID: 11034371
  • Nitcheu J, Bonduelle O, Combadiere C, Tefit M, Seilhean D, Mazier D, et al. Perforin-dependent brain-infiltrating cytotoxic CD8+ T lymphocytes mediate experimental cerebral malaria pathogenesis. J Immunol 2003; 170:2221 - 8; PMID: 12574396
  • Miyakoda M, Kimura D, Yuda M, Chinzei Y, Shibata Y, Honma K, et al. Malaria-specific and nonspecific activation of CD8+ T cells during blood stage of Plasmodium berghei infection. J Immunol 2008; 181:1420 - 8; PMID: 18606696
  • Boubou MI, Collette A, Voegtle D, Mazier D, Cazenave PA, Pied S. T cell response in malaria pathogenesis: selective increase in T cells carrying the TCR Vbeta8 during experimental cerebral malaria. Int Immunol 1999; 11:1553 - 62; http://dx.doi.org/10.1093/intimm/11.9.1553; PMID: 10464176
  • Rénia L, Potter SM, Mauduit M, Rosa DS, Kayibanda M, Deschemin JC, et al. Pathogenic T cells in cerebral malaria. Int J Parasitol 2006; 36:547 - 54; http://dx.doi.org/10.1016/j.ijpara.2006.02.007; PMID: 16600241
  • Jambou R, Combes V, Jambou MJ, Weksler BB, Couraud PO, Grau GE. Plasmodium falciparum adhesion on human brain microvascular endothelial cells involves transmigration-like cup formation and induces opening of intercellular junctions. PLoS Pathog 2010; 6:e1001021; http://dx.doi.org/10.1371/journal.ppat.1001021; PMID: 20686652
  • Cardoso FL, Brites D, Brito MA. Looking at the blood-brain barrier: molecular anatomy and possible investigation approaches. Brain Res Rev 2010; 64:328 - 63; http://dx.doi.org/10.1016/j.brainresrev.2010.05.003; PMID: 20685221
  • Neuwelt EA, Bauer B, Fahlke C, Fricker G, Iadecola C, Janigro D, et al. Engaging neuroscience to advance translational research in brain barrier biology. Nat Rev Neurosci 2011; 12:169 - 82; http://dx.doi.org/10.1038/nrn2995; PMID: 21331083
  • Levin BE, Magnan C, Dunn-Meynell A, Le FC. Metabolic sensing and the brain: who, what, where, and how?. Endocrinology 2011; 152:2552 - 7; http://dx.doi.org/10.1210/en.2011-0194; PMID: 21521751
  • Miller DW. Immunobiology of the blood-brain barrier. J Neurovirol 1999; 5:570 - 8; http://dx.doi.org/10.3109/13550289909021286; PMID: 10602398
  • Maegraith B, Fletcher A. The pathogenesis of mammalian malaria. Adv Parasitol 1972; 10:49 - 75; http://dx.doi.org/10.1016/S0065-308X(08)60172-4; PMID: 4626184
  • Adams S, Brown H, Turner GD. Breaking down the blood-brain barrier: signaling a path to cerebral malaria?. Trends Parasitol 2002; 18:360 - 6; http://dx.doi.org/10.1016/S1471-4922(02)02353-X; PMID: 12377286
  • Medana IM, Turner GD. Human cerebral malaria and the blood-brain barrier. Int J Parasitol 2006; 36:555 - 68; http://dx.doi.org/10.1016/j.ijpara.2006.02.004; PMID: 16616145
  • Grab DJ, Chakravorty SJ, van der Heyde HC, Stins MF. How can microbial interactions with the blood brain barrier modulate astroglial and neuronal function?. Cell Microbiol 2011; 13:1470 - 8; http://dx.doi.org/10.1111/j.1462-5822.2011.01661.x; PMID: 21824246
  • Warrell DA, Looareesuwan S, Phillips RE, White NJ, Warrell MJ, Chapel HM, et al. Function of the blood-cerebrospinal fluid barrier in human cerebral malaria: rejection of the permeability hypothesis. Am J Trop Med Hyg 1986; 35:882 - 9; PMID: 2429567
  • Brown HC, Chau TT, Mai NT, Day NP, Sinh DX, White NJ, et al. Blood-brain barrier function in cerebral malaria and CNS infections in Vietnam. Neurology 2000; 55:104 - 11; PMID: 10891914
  • Brown H, Rogerson S, Taylor T, Tembo M, Mwenechanya J, Molyneux M, et al. Blood-brain barrier function in cerebral malaria in Malawian children. Am J Trop Med Hyg 2001; 64:207 - 13; PMID: 11442219
  • Chapel HM, Warrell DA, Looareesuwan S, White NJ, Phillips RE, Warrell MJ, et al. Intrathecal immunoglobulin synthesis in cerebral malaria. Clin Exp Immunol 1987; 67:524 - 30; PMID: 3301098
  • Turner GDH, Morrison H, Jones M, Davis TME, Looareesuwan S, Buley ID, et al. An immunohistochemical study of the pathology of fatal malaria - Evidence for widespread endothelial activation and a potential role for intercellular adhesion molecule-1 in cerebral sequestration. Am J Pathol 1994; 145:1057 - 69; PMID: 7526692
  • Combes V, El-Assaad F, Faille D, Jambou R, Hunt NH, Grau GE. Microvesiculation and cell interactions at the brain-endothelial interface in cerebral malaria pathogenesis. Prog Neurobiol 2010; 91:140 - 51; http://dx.doi.org/10.1016/j.pneurobio.2010.01.007; PMID: 20117166
  • Larkin D, de LB, Jenkins PV, Bunn J, Craig AG, Terraube V, et al. Severe Plasmodium falciparum malaria is associated with circulating ultra-large von Willebrand multimers and ADAMTS13 inhibition. PLoS Pathog 2009; 5:e1000349; http://dx.doi.org/10.1371/journal.ppat.1000349; PMID: 19300493
  • Brown H, Hien TT, Day N, Mai NT, Chuong LV, Chau TT, et al. Evidence of blood-brain barrier dysfunction in human cerebral malaria. Neuropathol Appl Neurobiol 1999; 25:331 - 40; http://dx.doi.org/10.1046/j.1365-2990.1999.00188.x; PMID: 10476050
  • Deininger MH, Kremsner PG, Meyermann R, Schluesener H. Macrophages/microglial cells in patients with cerebral malaria. Eur Cytokine Netw 2002; 13:173 - 85; PMID: 12101073
  • Newton CRJ, Peshu N, Kendall B, Kirkham FJ, Sowunmi A, Waruiru C, et al. Brain swelling and ischaemia in Kenyans with cerebral malaria. Arch Dis Child 1994; 70:281 - 7; http://dx.doi.org/10.1136/adc.70.4.281; PMID: 8185359
  • Patankar TF, Karnad DR, Shetty PG, Desai AP, Prasad SR. Adult cerebral malaria: prognostic importance of imaging findings and correlation with postmortem findings. Radiology 2002; 224:811 - 6; http://dx.doi.org/10.1148/radiol.2243010588; PMID: 12202719
  • Medana IM, Day NP, Sachanonta N, Mai NT, Dondorp AM, Pongponrat E, et al. Coma in fatal adult human malaria is not caused by cerebral oedema. Malar J 2011; 10:267; http://dx.doi.org/10.1186/1475-2875-10-267; PMID: 21923924
  • Shinjo K, Tsuda S, Hayami T, Asahi T, Kawaharada H. Increase in permeability of human endothelial cell monolayer by recombinant human lymphotoxin. Biochem Biophys Res Commun 1989; 162:1431 - 7; http://dx.doi.org/10.1016/0006-291X(89)90834-6; PMID: 2788411
  • Royall JA, Berkow RL, Beckman JS, Cunningham MK, Matalon S, Freeman BA. Tumor necrosis factor and interleukin 1 alpha increase vascular endothelial permeability. Am J Physiol 1989; 257:L399 - 410; PMID: 2610269
  • Mark KS, Miller DW. Increased permeability of primary cultured brain microvessel endothelial cell monolayers following TNF-alpha exposure. Life Sci 1999; 64:1941 - 53; http://dx.doi.org/10.1016/S0024-3205(99)00139-3; PMID: 10353592
  • Gloor SM, Weber A, Adachi N, Frei K. Interleukin-1 modulates protein tyrosine phosphatase activity and permeability of brain endothelial cells. Biochem Biophys Res Commun 1997; 239:804 - 9; http://dx.doi.org/10.1006/bbrc.1997.7557; PMID: 9367850
  • Clark IA, Cowden WB, Rockett KA. Nitric oxide and cerebral malaria. Lancet 1993; 341:632 - 3; http://dx.doi.org/10.1016/0140-6736(93)90393-U; PMID: 7679766
  • Pino P, Vouldoukis I, Dugas N, Hassani-Loppion G, Dugas B, Mazier D. Redox-Dependent Apoptosis in Human Endothelial Cells after Adhesion of Plasmodium falciparum-Infected Erythrocytes. Ann N Y Acad Sci 2003; 1010:582 - 6; http://dx.doi.org/10.1196/annals.1299.109; PMID: 15033796
  • Taoufiq Z, Pino P, Dugas N, Conti M, Tefit M, Mazier D, et al. Transient supplementation of superoxide dismutase protects endothelial cells against Plasmodium falciparum-induced oxidative stress. Mol Biochem Parasitol 2006; 150:166 - 73; http://dx.doi.org/10.1016/j.molbiopara.2006.07.008; PMID: 16930739
  • Lopansri BK, Anstey NM, Weinberg JB, Stoddard GJ, Hobbs MR, Levesque MC, et al. Low plasma arginine concentrations in children with cerebral malaria and decreased nitric oxide production. Lancet 2003; 361:676 - 8; http://dx.doi.org/10.1016/S0140-6736(03)12564-0; PMID: 12606182
  • Weinberg JB, Lopansri BK, Mwaikambo E, Granger DL. Arginine, nitric oxide, carbon monoxide, and endothelial function in severe malaria. Curr Opin Infect Dis 2008; 21:468 - 75; http://dx.doi.org/10.1097/QCO.0b013e32830ef5cf; PMID: 18725795
  • Clark IA, Awburn MM, Whitten RO, Harper CG, Liomba NG, Molyneux ME, et al. Tissue distribution of migration inhibitory factor and inducible nitric oxide synthase in falciparum malaria and sepsis in African children. Malar J 2003; 2:6; http://dx.doi.org/10.1186/1475-2875-2-6; PMID: 12716455
  • Maneerat Y, Viriyavejakul P, Punpoowong B, Jones M, Wilairatana P, Pongponratn E, et al. Inducible nitric oxide synthase expression is increased in the brain in fatal cerebral malaria. Histopathology 2000; 37:269 - 77; http://dx.doi.org/10.1046/j.1365-2559.2000.00989.x; PMID: 10971704
  • Wong D, Dorovini-Zis K, Vincent SR. Cytokines, nitric oxide, and cGMP modulate the permeability of an in vitro model of the human blood-brain barrier. Exp Neurol 2004; 190:446; http://dx.doi.org/10.1016/j.expneurol.2004.08.008; PMID: 15530883
  • Udeinya IJ, Akogyeram CO. Induction of adhesiveness in human endothelial cells by Plasmodium falciparum-infected erythrocytes. Am J Trop Med Hyg 1993; 48:488 - 95; PMID: 8480856
  • Treeratanapiboon L, Psathaki K, Wegener J, Looareesuwan S, Galla HJ, Udomsangpetch R. In vitro study of malaria parasite induced disruption of blood-brain barrier. Biochem Biophys Res Commun 2005; 335:810 - 8; http://dx.doi.org/10.1016/j.bbrc.2005.07.151; PMID: 16105659
  • Brown H, Turner GDH, Rogerson SJ, Tembo M, Mwenechanya J, Molyneux ME, et al. Cytokine expression in the brain in human cerebral malaria. J Infect Dis 1999; 180:1742 - 6; http://dx.doi.org/10.1086/315078; PMID: 10515846
  • Tripathi AK, Sullivan DJ, Stins MF. Plasmodium falciparum-infected erythrocytes increase intercellular adhesion molecule 1 expression on brain endothelium through NF-kappaB. Infect Immun 2006; 74:3262 - 70; http://dx.doi.org/10.1128/IAI.01625-05; PMID: 16714553
  • Tripathi AK, Sha W, Shulaev V, Stins MF, Sullivan DJ Jr. Plasmodium falciparum-infected erythrocytes induce NF-kappaB regulated inflammatory pathways in human cerebral endothelium. Blood 2009; 114:4243 - 52; http://dx.doi.org/10.1182/blood-2009-06-226415; PMID: 19713460
  • Zougbédé S, Miller F, Ravassard P, Rebollo A, Ciceron L, Couraud PO, et al. Metabolic acidosis induced by Plasmodium falciparum intraerythrocytic stages alters blood-brain barrier integrity. J Cereb Blood Flow Metab 2011; 31:514 - 26; http://dx.doi.org/10.1038/jcbfm.2010.121; PMID: 20683453
  • Maneerat Y, Pongponratn E, Viriyavejakul P, Punpoowong B, Looareesuwan S, Udomsangpetch R. Cytokines associated with pathology in the brain tissue of fatal malaria. Southeast Asian J Trop Med Public Health 1999; 30:643 - 9; PMID: 10928354
  • Baruch DI, Pasloske BL, Singh HB, Bi X, Ma XC, Feldman M, et al. Cloning the Plasmodium falciparum gene encoding PfEMP1, a malarial variant antigen and adherence receptor on the surface of parasitized human erythrocytes. Cell 1995; 82:77 - 87; http://dx.doi.org/10.1016/0092-8674(95)90054-3; PMID: 7541722
  • Su XZ, Heatwole VM, Wertheimer SP, Guinet F, Herrfeldt JA, Peterson DS, et al. The large diverse gene family var encodes proteins involved in cytoadherence and antigenic variation of Plasmodium falciparum-infected erythrocytes. Cell 1995; 82:89 - 100; http://dx.doi.org/10.1016/0092-8674(95)90055-1; PMID: 7606788
  • Smith JD, Chitnis CE, Craig AG, Roberts DJ, Hudson-Taylor DE, Peterson DS, et al. Switches in expression of Plasmodium falciparum var genes correlate with changes in antigenic and cytoadherent phenotypes of infected erythrocytes. Cell 1995; 82:101 - 10; http://dx.doi.org/10.1016/0092-8674(95)90056-X; PMID: 7606775
  • Pasloske BL, Howard RJ. Malaria, the red cell, and the endothelium. Annu Rev Med 1994; 45:283 - 95; http://dx.doi.org/10.1146/annurev.med.45.1.283; PMID: 8198384
  • Schofield L, Grau GE. Immunological processes in malaria pathogenesis. Nat Rev Immunol 2005; 5:722 - 35; http://dx.doi.org/10.1038/nri1686; PMID: 16138104
  • Biswas AK, Hafiz A, Banerjee B, Kim KS, Datta K, Chitnis CE. Plasmodium falciparum uses gC1qR/HABP1/p32 as a receptor to bind to vascular endothelium and for platelet-mediated clumping. PLoS Pathog 2007; 3:1271 - 80; http://dx.doi.org/10.1371/journal.ppat.0030130; PMID: 17907801
  • Hatabu T, Kawazu SI, Aikawa M, Kano S. Binding of Plasmodium falciparum-infected erythrocytes to the membrane-bound form of Fractalkine/CX3CL1. Proc Natl Acad Sci USA 2003; 100:15942 - 6; http://dx.doi.org/10.1073/pnas.2534560100; PMID: 14665693
  • Etienne-Manneville S, Manneville JB, Adamson P, Wilbourn B, Greenwood J, Couraud PO. ICAM-1-coupled cytoskeletal rearrangements and transendothelial lymphocyte migration involve intracellular calcium signaling in brain endothelial cell lines. J Immunol 2000; 165:3375 - 83; PMID: 10975856
  • Couty JP, Rampon C, Leveque M, Laran-Chich MP, Bourdoulous S, Greenwood J, et al. PECAM-1 engagement counteracts ICAM-1-induced signaling in brain vascular endothelial cells. J Neurochem 2007; 103:793 - 801; http://dx.doi.org/10.1111/j.1471-4159.2007.04782.x; PMID: 17662049
  • Pino P, Vouldoukis I, Kolb JP, Mahmoudi N, Desportes-Livage I, Bricaire F, et al. Plasmodium falciparum-Infected Erythrocyte Adhesion Induces Caspase Activation and Apoptosis in Human Endothelial Cells. J Infect Dis 2003; 187:1283 - 90; http://dx.doi.org/10.1086/373992; PMID: 12696008
  • Touré FS, Ouwe-Missi-Oukem-Boyer O, Bisvigou U, Moussa O, Rogier C, Pino P, et al. Apoptosis: a potential triggering mechanism of neurological manifestation in Plasmodium falciparum malaria. Parasite Immunol 2008; 30:47 - 51; PMID: 18086016
  • Wassmer SC, Moxon CA, Taylor T, Grau GE, Molyneux ME, Craig AG. Vascular endothelial cells cultured from patients with cerebral or uncomplicated malaria exhibit differential reactivity to TNF. Cell Microbiol 2011; 13:198 - 209; http://dx.doi.org/10.1111/j.1462-5822.2010.01528.x; PMID: 21029292
  • Siau A, Toure FS, Ouwe-Missi-Oukem-Boyer O, Ciceron L, Mahmoudi N, Vaquero C, et al. Whole-transcriptome analysis of Plasmodium falciparum field isolates: identification of new pathogenicity factors. J Infect Dis 2007; 196:1603 - 12; http://dx.doi.org/10.1086/522012; PMID: 18008243
  • Pino P, Taoufiq Z, Nitcheu J, Vouldoukis I, Mazier D. Blood-brain barrier breakdown during cerebral malaria: suicide or murder?. Thromb Haemost 2005; 94:336 - 40; PMID: 16113823
  • Faille D, El-Assaad F, Alessi MC, Fusai T, Combes V, Grau GE. Platelet-endothelial cell interactions in cerebral malaria: the end of a cordial understanding. Thromb Haemost 2009; 102:1093 - 102; PMID: 19967139
  • Deininger MH, Kremsner PG, Meyermann R, Schluesener HJ. Differential cellular accumulation of transforming growth factor-beta1, -beta2, and -beta3 in brains of patients who died with cerebral malaria. J Infect Dis 2000; 181:2111 - 5; http://dx.doi.org/10.1086/315493; PMID: 10837206
  • Rogerson SJ, Grau GE, Hunt NH. The microcirculation in severe malaria. Microcirculation 2004; 11:559 - 76; http://dx.doi.org/10.1080/10739680490503311; PMID: 15513866
  • Dondorp AM, Pongponratn E, White NJ. Reduced microcirculatory flow in severe falciparum malaria: pathophysiology and electron-microscopic pathology. Acta Trop 2004; 89:309 - 17; http://dx.doi.org/10.1016/j.actatropica.2003.10.004; PMID: 14744557
  • Migasena P, Maegraith BG. The movement of the dye (disulphine blue) from blood into brain tissue examined by dye method in normal and Plasmodium berghei-infected mice. Med J Malaya 1968; 22:252; PMID: 4234391
  • Ma N, Madigan MC, Chan-Ling T, Hunt NH. Compromised blood-nerve barrier, astrogliosis, and myelin disruption in optic nerves during fatal murine cerebral malaria. Glia 1997; 19:135 - 51; http://dx.doi.org/10.1002/(SICI)1098-1136(199702)19:2<135::AID-GLIA5>3.0.CO;2-#; PMID: 9034830
  • Thumwood CM, Hunt NH, Clark IA, Cowden WB. Breakdown of the blood-brain barrier in murine cerebral malaria. Parasitology 1988; 96:579 - 89; http://dx.doi.org/10.1017/S0031182000080203; PMID: 2457201
  • van der Heyde HC, Bauer PR, Sun G, Chang WL, Yin L, Fuseler J, et al. Assessing vascular permeability during experimental cerebral malaria by a Radiolabeled Monoclonal Antibody Technique. Infect Immun 2001; 69:3460 - 5; http://dx.doi.org/10.1128/IAI.69.5.3460-3465.2001; PMID: 11292776
  • Neill AL, Chan-Ling T, Hunt NH. Comparisons between microvascular changes in cerebral and non-cerebral malaria in mice, using the retinal whole-mount technique. Parasitology 1993; 107:477 - 87; http://dx.doi.org/10.1017/S0031182000068050; PMID: 8295787
  • Chang-Ling T, Neill AL, Hunt NH. Early microvascular changes in murine cerebral malaria detected in retinal wholemounts. Am J Pathol 1992; 140:1121 - 30; PMID: 1374593
  • Medana IM, Hunt NH, Chan-Ling T. Early activation of microglia in the pathogenesis of fatal murine cerebral malaria. Glia 1997; 19:91 - 103; http://dx.doi.org/10.1002/(SICI)1098-1136(199702)19:2<91::AID-GLIA1>3.0.CO;2-C; PMID: 9034826
  • Penet MF, Viola A, Confort-Gouny S, Le Fur Y, Duhamel G, Kober F, et al. Imaging experimental cerebral malaria in vivo: significant role of ischemic brain oedema. J Neurosci 2005; 25:7352 - 8; http://dx.doi.org/10.1523/JNEUROSCI.1002-05.2005; PMID: 16093385
  • Penet MF, Kober F, Confort-Gouny S, Le Fur Y, Dalmasso C, Coltel N, et al. Magnetic resonance spectroscopy reveals an impaired brain metabolic profile in mice resistant to cerebral malaria infected with Plasmodium berghei ANKA. J Biol Chem 2007; 282:14505 - 14; http://dx.doi.org/10.1074/jbc.M608035200; PMID: 17369263
  • Ammapawong S, Combes V, Hunt NH, Radford J, Chan-Ling T, Pongponratn E, et al. Quantitation of brain oedema and localisation of aquaporin 4 expression in relation to susceptibility to experimental cerebral malaria Int J. Clin Exp Pathol 2011; 4:566 - 74
  • Huynh HK, Dorovini-Zis K. Effects of interferon-gamma on primary cultures of human brain microvessel endothelial cells. Am J Pathol 1993; 142:1265 - 78; PMID: 8475997
  • Bauer PR, van der Heyde HC, Sun G, Specian RD, Granger DN. Regulation of endothelial cell adhesion molecule expression in an experimental model of cerebral malaria. Microcirculation 2002; 9:463 - 70; PMID: 12483543
  • von Zur Muhlen C, Sibson NR, Peter K, Campbell SJ, Wilainam P, Grau GE, et al. A contrast agent recognizing activated platelets reveals murine cerebral malaria pathology undetectable by conventional MRI. J Clin Invest 2008; 118:1198 - 207; PMID: 18274670
  • Lou J, Donati YR, Juillard P, Giroud C, Vesin C, Mili N, et al. Platelets play an important role in TNF-induced microvascular endothelial cell pathology. Am J Pathol 1997; 151:1397 - 405; PMID: 9358766
  • Cabrales P, Zanini GM, Meays D, Frangos JA, Carvalho LJ. Nitric Oxide Protection Against Murine Cerebral Malaria Is Associated With Improved Cerebral Microcirculatory Physiology. J Infect Dis 2011; 203:1454 - 63; http://dx.doi.org/10.1093/infdis/jir058; PMID: 21415018
  • Gramaglia I, Sobolewski P, Meays D, Contreras R, Nolan JP, Frangos JA, et al. Low nitric oxide bioavailability contributes to the genesis of experimental cerebral malaria. Nat Med 2006; 12:1417 - 22; http://dx.doi.org/10.1038/nm1499; PMID: 17099710
  • Potter S, Chan-Ling T, Ball HJ, Mansour H, Mitchell A, Maluish L, et al. Perforin mediated apoptosis of cerebral microvascular endothelial cells during experimental cerebral malaria. Int J Parasitol 2006; 36:485 - 96; http://dx.doi.org/10.1016/j.ijpara.2005.12.005; PMID: 16500656
  • Suidan GL, McDole JR, Chen Y, Pirko I, Johnson AJ. Induction of blood brain barrier tight junction protein alterations by CD8 T cells. PLoS ONE 2008; 3:e3037; http://dx.doi.org/10.1371/journal.pone.0003037; PMID: 18725947