R e vie w

2 downloads 0 Views 412KB Size Report
duces the type of brain edema seen in pediatric rather than adult ...... Coma in fatal adult human malaria is not caused by cerebral oedema. Malar. J. 10, 267.
Georges Emile Raymond Grau*1,2 & Alister Gordon Craig3 Vascular Immunology Unit, Department of Pathology, Sydney Medical School, The University of Sydney, Camperdown NSW 2042, Australia La Jolla Infectious Disease Institute, San Diego, CA 92109, USA 3 Malawi Liverpool School of Tropical Medicine, Liverpool, UK *Author for correspondence: [email protected] 1

2

What is cerebral malaria?

adhesive interactions between IE and host cells, including endothelium, but have also ranged from host genetic studies to clinical measurements of a wide range of systemic and local effectors. So, while we do not fully understand the pathology of CM and suspect that it may have multiple etiologies, we do know that it has some differences to, and some overlaps with, other brain inflammatory diseases and we have information about some of the potential contributions from the parasite and the host that could lead to CM.

A

ut

ho

rP

Cerebral malaria (CM) forms part of the spectrum of severe malaria, with a case fatality rate ranging from 15% in adults in southeast Asia [1] to 8.5% in children in Africa [2] . Clinical signs of acidosis carry a higher risk of death but nevertheless CM accounts for a significant proportion of malaria mortality, as well as the potential for neurological deficits in survivors. The standard clinical definition of CM centers on a state of unarousable coma partnered with the presence of malaria infected red blood cells in the peripheral circulation and a lack of other potential causes of coma such as other infections or hypoglycemia (for a full definition see [3–5]). More recently ophthalmic observations of retinopathy have been added to this definition in both adults and children to increase the specificity of the clinical diagnosis [6,7] . Most observations of the pathophysiology of disease come from postmortem observations of Plasmodium falciparum (Pf) infections, which are thought to account for the vast majority of CM cases, and show a common feature of vascular sequestration of infected erythrocytes (IE) in the brain [8] . There are also some differences, particularly between CM in adults and children, broadly separable into a ‘pure’ sequestration pattern and IE sequestration with variable (and moderate) vascular pathology. The latter varies from the accumulation of proinflammatory cells such as leukocytes and platelets to localized vascular damage (e.g., vessels partially denuded of endothelium) [9] . With the hallmark of IE sequestration for CM (albeit based on postmortem studies), investigations into the pathology of disease have looked at the

ro

of

Cerebral malaria is one of a number of clinical syndromes associated with infection by human malaria parasites of the genus Plasmodium. The etiology of cerebral malaria derives from sequestration of parasitized red cells in brain microvasculature and is thought to be enhanced by the proinflammatory status of the host and virulence characteristics of the infecting parasite variant. In this article we examine the range of factors thought to influence the development of Plasmodium falciparum cerebral malaria in humans and review the evidence to support their role.

Review

Future Microbiology

Cerebral malaria pathogenesis: revisiting parasite and host contributions

10.2217/FMB.11.155 © 2012 Future Medicine Ltd

Parasite contributions PfEMP1 & cytoadherence

When Pf merozoites invade red blood cells they remodel the host cell extensively through changes to the cytoskeleton and insertion of parasite-derived proteins into and onto the erythrocyte membrane (for review see [10]). One such protein, thought to be a major virulence factor in Pf, is the major variant surface antigen PfEMP1. Owing to its expression on the surface of the IE, this protein must undergo antigenic variation to escape host defense mechanisms and a complex mechanism exists to support this [11] . This variable presentation of PfEMP1 proteins to the host results in differences between parasite antigenic variants in the immune response that they provoke and in the repertoire of host receptors that erythrocytes infected with different variants can bind to (as well as the affinity of this binding) [12] . PfEMP1 is encoded by a family of var genes that can be characterized by sequence motifs at the 5´ promoter region and within the first Future Microbiol. (2012) 7(2), 1–xxx

Keywords keyword n keyword n keyword

n

part of

ISSN 1746-0913

1

Review

Grau & Craig

effect of these IE on the host. This is further complicated by findings that the rigidity of IE itself, caused most likely through the parasitederived modifications to the erythrocyte, is also associated with severe disease [25,26] . How might IE sequestration lead to pathology? Earlier considerations of this issue divided into two main hypotheses: mechanical obstruction of vessels and inflammation. More recently the boundaries between these categories have become blurred and it seems most likely that combinations of both of these processes are responsible for disease, and support for mechanical blockage being a significant factor has come recently from elegant observations of the mucosal microvasculature performed by Dondorp et al. [27] . What has become clear is that the binding interaction between IE and host endothelium is not a passive process and the engagement and adjacency for these cell types results in the activation of a range of activities ascribed to the binding interaction itself, biomechanical stimuli and the localized release of soluble or membrane-bound mediators [28–31] . Two major categories of endothelial ‘damage’ have been reported, namely a breakdown of the barrier function of the specialized blood–brain barrier (BBB) and apoptosis of ECs. BBB integrity has been shown in postmortem studies to be subtly disturbed during severe malaria resulting in some leakage from cerebral vessels [32] and further studies in vitro have supported the role of IE as well as host effector cells in this process [33–35] . Much of the latter work has used measurement of transendothelial electrical resistance in EC/IE coculture models, identifying transcriptional changes caused through this interaction [33] and, in some cases, alterations to the junctional proteins (e.g., ZO‑1 and occludin) at the EC–EC interface [36,37] . In terms of apoptosis, patient isolates have been shown to have variable apoptosis-inducing abilities, with a weak relationship between an increased induction of EC apoptosis by isolates taken from children with neurological manifestations [38] . In vitro work also supports the hypothesis that vascular damage may be caused by apoptotic destruction of the ECs, particularly in combination with platelets or microparticles [39–41] . It is possible that a balance exists between the triggering of apoptosis by the engagement of host receptors during cytoadherence and IE–EC apposition, and the ability of the IE to modulate this response [30] , with some of the highly localized vascular damage seen on postmortem of people dying of CM as micro-hemorrhages

rP

ro

of

adhesive (DBL) domain. Analysis of these sequence tags has identified associations of specific subsets of var genes with severe disease [13–17] , supporting the idea that PfEMP1 is a virulence factor. For example, Warimwe et al. showed a positive correlation between Group A-like/cys2 (or Ups-A) var gene expression and impaired consciousness, reinforcing a number of studies that had suggested associations between Ups-A var gene expression in the invading isolate and severe malaria. This sequence variant type is also selected from parasite populations using sera from semi-immune children [18,19] , the same category that shows greater susceptibility to CM. The relationship is not exact and many Ups-A/cys2 type infections do not lead to CM or even other aspects of severe malaria, which probably reflects the requirement of particular host–pathogen combinations to drive specific disease patterns. However, in considering potential parasite pathogenic processes, the ability of IE to undergo a range of adhesive interactions with host cells has been a major area of study. Binding interactions between IE and host cells can be divided into three main categories: Cytoadherence (interaction with endothelial cells [ECs] or, in placental malaria, with the syncytiotrophoblast cells of the placenta);

n

Rosetting (interaction with uninfected erythrocytes);

ho

n

Clumping (interaction with other IE via platelet bridges).

n

A

ut

The molecular basis for these interactions have largely been described (for review see [12]) although there is clearly scope for the identification of further host receptors. The picture emerging is a complex one with over 12 host receptors available to the parasite through its repertoire of PfEMP1 protein variants, used in different permutations and with varying affinities. Several studies have attempted to correlate specific binding activity with clinical outcomes through testing binding phenotypes of patient isolates. Rosetting and clumping have both been variably associated with severe malaria [20–23] but results for cytoadherence to endothelium have been variable with only one recent study showing a statistically significant association between higher binding to ICAM‑1 under flow assay conditions and CM [24] . Nor do these binding experiments explain how the sequestration of IE in the brain leads to CM and an explanation of this awaits further work on the processes leading to differential cerebral IE sequestration and the

2

Future Microbiol. (2012) 7(2)

future science group

Cerebral malaria pathogenesis: revisiting parasite & host contributions

potentially being caused through a failure to control EC apoptosis by the parasite.

pathways such as apoptosis, BBB integrity and increased receptor expression.

Cytoadherence-related signaling pathways

Systemic & localized inflammation

ro

of

As well as interactions with endothelium, IE are also able to release mediators directly causing host effector cells (and ECs) to release a range of pro- and anti-inflammatory cytokines. Pf glycosylphosphatidylinositol is released during schizont rupture and is able to stimulate macrophages to release TNF and IL‑1 [50] , contributing to the systemic proinflammatory response seen during malaria infection. Pf glycosylphosphatidylinositol has been shown to increase expression of ICAM‑1, potentially increasing the recruitment of IE to the cerebral vasculature [51] , as well as inducing nitric oxide (NO) production [52] , although the link between NO and CM pathology is less clear. It is also able to induce limited apoptosis in some tissues [53,54] , although these have not included brain. The other major parasite component capable of directly stimulating the host immune system is the malaria pigment known as hemozoin (or hematin). Hemozoin is produced during the digestion of hemoglobin in the parasite digestive vacuole in order to detoxify the heme moiety and mediates a broad range of inflammatory and immunomodulatory activities (for review see [55]). The characteristic brown crystals of hemozoin are seen in a number of different phagocytic cells and appear to be able to persist despite the degradation pathways available within these cells. Hemozoin-containing monocytes and neutrophils have been found in greater numbers in adults dying of severe malaria compared with survivors [56] as well as children with CM [9,57] . Research on the role of hemozoin in inflammation has been complicated by the heterogeneity of experimental conditions, not the least being the source of hemozoin, either synthesized or natural. However it seems clear that it is able to induce the production of proinflammatory cytokines from a range of host cells contributing to the immunopathology of malaria, including CM. Hemozoin is also thought to be able to influence the adaptive immunity [58,59] , altering the ability of the host to make an effective response to infection. It is also involved in the inhibition of erythropoiesis and thereby to malarial anemia [60] . It has recently been suggested that the host fibrinogen bound to hemozoin is principally responsible for at least some of the proinflammatory stimulation seen [61] . A direct link with CM is harder to discern. The systemic proinflammatory response seen with

A

ut

ho

rP

Further evidence that the IE/EC interaction is not a passive one comes from various reports demonstrating the activation of signaling pathways. Receptor clustering during adhesive events is a common pathway for activation of signaling cascades and cytoadherence is able to mimic these events. IE binding via CD36 activates a Src-family kinase dependent MAP kinase pathway and this Src-family kinase activity is required for efficient cytoadherence such that treatment of EC with specific inhibitors was able to reduce IE binding [42] . The same group also showed that ectophosphorylation of CD36 itself, through a Src-family kinase/ alkaline phosphatase pathway, also controlled adhesion to this receptor [43] . This information has subsequently been used to support clinical intervention trials with the alkaline phosphatase inhibitor levamisole, which are currently underway [44] . Cerebral endothelium has low levels of CD36 that would not normally support IE adhesion so its relevance to CM has been questioned. In addition, higher levels of binding to CD36 have not been associated with CM, either being linked to non-CM severe malaria in one study [45] and to uncomplicated malaria in another [24] (exref #22). However, studies have shown that platelets and microparticles are able to act as a bridge to support adhesion to cerebral EC [46,47] and even to transfer CD36 to these cells [34] . Thus it is possible that CD36 may sometimes be available for adhesion in the brain, or alternatively supports efficient exponential parasite multiplication during the asexual growth phase indirectly contributing to subsequent pathology. ICAM‑1 is available for IE cytoadherence in the brain in the context of the widespread endothelial activation seen in CM and significant colocalization of this receptor (as well as CD36 and E‑selectin) has been seen with IE sequestration in adults [48] . IE are also able to activate host signaling by binding to ICAM‑1, via the MAP kinases ERK1/2, JNK and p38 [49] . The nuclear transcription factor NF‑kB is also implicated in cerebral cytoadherence, as demonstrated by transcriptional ana­lysis of IE/brain EC coculture showing the differential expression of pathways controlled by NF-kB [31] . The full impact of these signaling changes on CM is not known but experiments have implicated a number of pathogenic

Review

future science group

www.futuremedicine.com

3

Review

Grau & Craig

Tie2 signaling system [83] . Ang1:Ang2 ratios have recently been identified as one of the best biomarkers for CM in African children [84] . In eCM, using the Plasmodium berghei ANKA (PbA) mouse model [85] , TNF [86] and IFN‑g [87] have been identified as important mediators of disease, and LTas being possibly the principal mediator responsible for the triggering of neuropathology [88] . Using an intervention approach, a recent confirmation of the role of cytokines in this model has been provided by the demonstration that CM can be induced in CM-resistant mice by inducing the endogenous secretion of proinflammatory cytokines. This was achieved by injections of CpG [89] . IFN‑g has clearly been shown to be central in eCM pathogenesis [87] , and several pathways can be triggered by this cytokine, notably IP‑10/CXCL10 [90] .A complication with many of these studies (in human and mouse systems) is the balance between protection and pathology elicited by the cytokine response. For example, experiments using the mouse model of eCM have determined that early production of this cytokine in the infection can be protective rather than deleterious [91] . Other host mediators of importance in eCM pathogenesis include histamine [92] , via its H(3) receptor [93] , complement, particularly C5a [94] , endothelin [95] and HO‑1 (reviewed in [96]). Aside from cytokines and noncytokine mediators, coagulation factors also participate in the triggering of pathology of eCM, as reviewed in [97] .

rP

ro

of

hemozoin would directly support cytoadherence-mediated pathology and some evidence exists to support activation of matrix metalloproteinases (e.g., MMP‑9 [62]) that could cause vascular damage and induce morphological changes to endothelium [63] . The modulation of host protective responses could also have an indirect action via parasite multiplication and retention of parasite material in cerebral vessels. Endothelial activation appears to be strongly associated with CM [48,64] and the basis for this is likely to be multifactorial. Von Willebrand factor (vWF) is raised in CM [65,66] at the same time that ADAMTS13 activity, which is responsible for degrading the bioactive ultra-large vWF multimers expressed on the EC surface, is reduced [67–69] . Part of the mechanism responsible for this association may be the cytoadherence of IE to platelets decorating the ultra-large vWF expressed on the EC [47] , including brain tissues. Release of ultra-large vWF requires activation and mobilization of the Weibel-Palade bodies (for review see [70]), which can occur through several mediators including histamine. The malaria parasite Pf produces a protein, PfTCTP, which is a homolog for the mammalian histamine releasing factor and can induce histamine release from basophils [71] .

ho

Host contributions

For the sake of clarity, we will address findings in human CM (hCM) and those in experimental CM (eCM), separately.

Effector cells

More than half a century after Brian Maegraith’s original suggestions [72,73] that endogenous mediators have a pathogenic role in severe malaria, a body of evidence suggests that cytokines are involved in CM pathogenesis: reviewed in [74–76] . In hCM, high levels of such proinflammatory cytokines have been reported for more than two decades [77–79] , and confirmed by numerous groups [80] even if some reports suggest that there is no association with severity and that endothelial molecules might be more reliable [64] . In the human disease, at least in its in vitro modeling, attention has been drawn on the role of TF [81] and more recently vWF [47] . The latter seems crucial in the early steps of Pf cytoadherence, in a dynamic flow chamber setting [82] . In addition to the release of vWF, the activation of the Weibel-Palade body also influences local EC activation through release of Ang2 via the

In hCM, only a few intravascular leukocytes are seen in brain sections of CM2 patients [9,98] , but the potential role of these cells remains unknown. HIV‑1 positive subjects are at risk of developing severe malaria (reviewed in [99]) but, while it has been proposed that AIDS and CM do not influence each other [100] , the relationship between these two syndromes is exceedingly complex, as discussed in [101,102] . In eCM, the T‑cell dependency of the neurological syndrome is clear, involving both CD4 + [103,104] and CD8 + [105,106] T cells (reviewed in [74,107]). Histopathologically, in eCM the presence of leukocytes is more conspicuous than in hCM, but no CD8 T‑cells are visible locally, yet CD8 depletion prevents eCM [97] . The issue of Tregs in eCM has been addressed by Engwerda’s team [108] , who showed that CD4 + CD25 + regulatory T cells suppress CD4 + T‑cell function

A

ut

Mediators: cytokines & coagulation factors

4

Future Microbiol. (2012) 7(2)

Leukocytes

future science group

Cerebral malaria pathogenesis: revisiting parasite & host contributions

A

ut

ho

rP

In African patients with hCM, platelets have been found to accumulate in brain vessels, but not in cases of severe malarial anemia or coma of other causes [98] . This intravascular accumulation has been recently confirmed at the level of CM-associated retinopathy [113]. In eCM, platelets are found to accumulate in organs before these show lesions, as revealed by radiolabeled platelet localization experiments [114] , and intervention experiments have demonstrated that they are involved in pathology (reviewed in [115]). Furthermore, intravascular platelets are detectable by the laser-induced breakdown spectroscopy technology even before microvascular dysfunction can be detected by MRI [116] . These in vivo data have been analyzed in further detail in vitro, using mouse EC [117] , as well as human [39] EC, cocultures, notably disclosing the importance of the release of TGF‑b1 by platelets [118] . More recently, using an in  vitro model of human CM, it was found that platelets can trigger a number of EC genes [119] involved in inflammation and apoptosis, such as genes involved in chemokine, TREM1, cytokine, IL‑10, TGFb, death-receptor and apoptosis signaling.

of

Platelets

has been shown in  vitro, using human cells [126] . The effect of these direct interactions is amplified by the cascade of inflammatory reactions discussed in the ‘Mediators: cytokines & coagulation factors’ and ‘Effector cells’ sections. Associated with this, a number of endothelial functions become altered [34] , notably permeability, leading to brain edema, which can be mild in adult CM [127] and more pronounced in pediatric CM [128] . In this respect, eCM reproduces the type of brain edema seen in pediatric rather than adult hCM, as evidenced by MRI [129] , including at the eye level [130] and by quantitative immunohistochemistry [131] . The latter study showed a marked upregulation of aquaporin 4 on astrocytic foot processes in eCM, a finding that has recently been found marginally modified in hCM, in adults where brain edema does not appear to be the cause of death [132] . However, even if brain swelling is found in adult patients with CM, simple therapeutic measures against brain edema, such as mannitol do not protect against the neurological syndrome, and may even be harmful [133] . Therefore, fine mechanisms leading to brain edema need to be investigated in more depth. Angiogenic dysregulation in severe malaria has been substantiated by the demonstration of altered levels of VEGF, soluble VEGFR‑2 [134] , and Ang2 [135] . Howver, studies differ in recording an increase or decrease in VEGF, perhaps owing to differences in the clinical groups being considered. In specific comparisons of hCM from India, VEGF was reduced in plasma from patients with hCM compared with mild malaria cases and healthy controls, and further reduced in patients dying from hCM [136] . Yeo et al. have suggested that the increase in endothelial activation marked by increasing levels of Ang2 could be owing to reduced bioavailability of NO, which in turn may result from a reduction in its precursor, l‑arginine. It is possible that the sequestered IE may cause this reduction through the release of arginases on schizont rupture. They showed that endothelial function, measuring reperfusion by reactive hyperemiaperipheral arterial tonometry [137] , was reduced in severe malaria and subsequently demonstrated that this deficiency could be partially rectified by the infusion of l‑arginine [138] . Most likely is that a subtle balance between positive and negative regulation of endothelial functions contributes to the endothelial pathology of CM, as discussed elsewhere [139] . Interestingly, an association has been found between susceptibility to CM development and

ro

and inhibit the development of P. berghei-specific Th1 responses involved in eCM pathogenesis [109] . More recently, however, in 2010, Haque et al. demonstrated that in vivo expansion of Treg by using IL‑2/anti-IL‑2 complexes prevents eCM [110] . In African patients, Treg deficiency appears to be protective and Treg cells have been found to be increased in patients with hyperparasitemia, therefore immune (or inflammation-based) pathology is clearly important in human severe malaria, including hCM [111,112] . The involvement of monocytes in eCM has been reviewed [74] .

Review

ECs: both targets & effectors in CM

Severe malaria is characterized by a pleomorphic endothelial cell (EC) activation. While studying various aspects of EC changes in relation to tissue damage, we showed that IE, together with host cells, leads to profound endothelial alterations [120] . In turn, EC can trigger immunopathological changes, as detailed in [121] . Among others, we identified miRNAs able to regulate ECs that are modulated in murine CM but not in uncomplicated malaria [122] . A central feature of endothelial changes in CM is activation [123–125] , resulting from the direct contact with IE, as discussed in the ‘Parasite contributions’ section, or with leukocytes, as future science group

www.futuremedicine.com

5

Review

Grau & Craig

endothelial reactivity to cytokines [140] , as had been suggested in the CM model [141] . The endothelial hyper-reactivity notably included the production of membrane microparticles, which are found in dramatically high numbers in patients with hCM [40,41,142] and could be novel players of disease pathogenesis, as reviewed extensively in [120,143–145] . Repercussions on brain parenchyma Glial changes

ho

rP

ro

of

In hCM, alterations of glial cells have been studied in tissues from patients as well as in culture systems. As integral constituents of the BBB, astrocytes can show signs of dysfunction as a result of the IE sequestration. In patients with CM, several aspects of microglial activation have also been reviewed [146] . In vitro, soluble factors from Pf induce apoptosis in human brain vascular endothelial and neuroglia cells. More DNA fragmentation is found in brain EC than in neuroglia, suggesting that extended exposure to high levels of these soluble factors in circulation is critical [147] . In eCM, similar glial changes, involving both astrocytes and microglial cells have been reported. The potential consequences of glial activation, notably glial-derived proteinases, in structural damage of the CNS have been reviewed in detail [148] . Among the inflammatory mediators, selected chemokines are dramatically upregulated in microvessels and adjacent glial cells [149] .

early changes not only in the optic but also the trigeminal nerves. Cranial nerve injury was the earliest anatomic hallmark of the disease, visible even before brain edema became detectable [130] . The overproduction of IFN‑g, whose crucial role in ECM has been shown [87] , can increase expression of the heme enzyme indoleamine 2,3-dioxygenase in microvascular ECs. Raised indoleamine 2,3-dioxygenase levels leads to increased production of a range of biologically active metabolites that may be part of a tissue protective response. Damage to astrocytes may result in reduced production of the neuroprotectant kynurenic acid, leading to a decrease in its ratio relative to the neuroexcitotoxic quinolinic acid. This imbalance may contribute to some of the neurological signs of human and experimental CM, as reviewed in Hunt et al. [154] . Reactive astrocytes also can show expression of a molecule that was thought to be a neuronspecific, hypoxia-responsive and neuroprotective protein: neuroglobin was found to be present, aside from neurons, in reactive astroglia and in scar-forming astrocytes in various neuropathological conditions, including CM [155] . CM is a complex pathology that illustrates the impact and the role of endothelial, glial and neuronal cells in the process. The global role of the neurovascular unit, considered as a whole or as an organ is discussed elsewhere [Grau et al. Manuscript in preparation] .

Neuronal

A

ut

In hCM, immunohistochemistry has been used to identify a variety of cellular stress and injury responses in the brains of patients with fatal falciparum malaria. Oxidative stress predominated in the vicinity of vessels and hemorrhages. Some degree of DNA damage was found in the majority of malaria patients, but staining patterns suggest considerable stress response and reversible neuronal injury [150] . Both axonal and astrocytic injury markers, the tau protein and S‑100B, respectively, have been found in elevated concentrations in CSF samples from CM patients [151] . A disruption of axonal transport was further demonstrated by detection of µ- and m‑calpain, the specific inhibitor calpastatin, in postmortem brain tissue from patients who died from severe malaria [152] . The first evidence for a loss of axonal viability in eCM and the first demonstration of optic nerve involvement was provided by Ma et al. [153] . Recently, MRI in this model revealed very

6

Future Microbiol. (2012) 7(2)

Behavioral/cognitive changes

The development of neurological sequelae after hCM has long been recognized and have been recently characterized in detail. It was found that Kenyan children have neurocognitive impairments that are evident as long as 9 years later [156,157] . Potential cognitive rehabilitation solutions such as cognitive exercises, environmental enrichment, nutritional supplementation, physical therapy and speech therapy have been proposed [158] . In Ugandan children, a computerized cognitive training program 3  months after severe malaria had an immediate effect on cognitive outcomes but did not affect academic skills or behavior [159] . In addition to previously described neurological and cognitive sequelae, severe behavior problems may follow CM in children, including ADHD. In another study in Ugandan children, observed differences in patterns of sequelae may be owing to different pathogenic mechanisms, brain regions affected or extent of injury [160] . In eCM, inflammatory changes in the CNS are also associated with behavioral impairment future science group

Cerebral malaria pathogenesis: revisiting parasite & host contributions

Conclusion

Financial & competing interests disclosure The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties. No writing assistance was utilized in the production of this manuscript.

ro

It seems likely that the pathologies underlying CM in humans are highly variable and reflect a range of attributes, including parasite virulence, host susceptibility and comorbidities ranging from malnutrition to coinfection.

Understanding the contribution of these components using all the tools at our disposal will be essential to the development of new antidisease interventions aimed at reducing the mortality and postinfection morbidity associated with cerebral malaria.

of

in P. berghei (strain ANKA)-infected mice 2010 [161] . As there is evidence that these behavioral changes may be due to – or at least associated with – oxidative stress, as assessed by elevated levels of malondialdehyde and conjugated dienes in the brains of PbA-infected C57BL/6 mice with CM, antioxidant treatment was evaluated as adjunct therapy. PbA-infected C57BL/6 mice with additive antioxidants together with chloroquine at the first signs of CM prevented the development of persistent cognitive damage [162] .

Review

References

ut

ho

rP

Executive summary

1.

2.

3.

A

Papers of special note have been highlighted as: n of interest nn of considerable interest

Dondorp A, Nosten F, Stepniewska K, Day N, White N. Artesunate versus quinine for treatment of severe falciparum malaria: a randomised trial. Lancet 366, 717–725 (2005). Dondorp AM, Fanello CI, Hendriksen IC et al. Artesunate versus quinine in the treatment of severe falciparum malaria in African children (AQUAMAT): an open-label, randomised trial. Lancet 376, 1647–1657 (2010). Severe falciparum malaria. World Health Organization, Communicable Diseases Cluster. Trans. R. Soc. Trop. Med. Hyg. 94(Suppl. 1), S1–S90 (2000).

future science group

4.

Idro R, Jenkins NE, Newton CR. Pathogenesis, clinical features, and neurological outcome of cerebral malaria. Lancet Neurol. 4, 827–840 (2005).

9.

5.

WHO. Guidelines for the Treatment of Malaria (2nd Edition). The WHO, Geneva, Switzerland (2010).

10. Maier AG, Cooke BM, Cowman AF, Tilley

6.

7.

8.

Maude RJ, Beare NA, Abu Sayeed A et al. The spectrum of retinopathy in adults with Plasmodium falciparum malaria. Trans. R. Soc. Trop. Med. Hyg. 103, 665–671 (2009). Beare NA, Lewallen S, Taylor TE, Molyneux ME. Redefining cerebral malaria by including malaria retinopathy. Future Microbiol. 6, 349–355 (2011). MacPherson GG, Warrell MJ, White NJ, Looareesuwan S, Warrell DA. Human cerebral malaria. A quantitative ultrastructural ana­lysis of parasitized erythrocyte sequestration. Am. J. Pathol. 119, 385–401 (1985).

www.futuremedicine.com

Dorovini-Zis K, Schmidt K, Huynh H et al. The neuropathology of fatal cerebral malaria in malawian children. Am. J. Pathol. 178, 2146–2158 (2011). L. Malaria parasite proteins that remodel the host erythrocyte. Nat. Rev. Microbiol. 7, 341–354 (2009).

11. Dzikowski R, Deitsch KW. Genetics of

antigenic variation in Plasmodium falciparum. Curr. Genet. 55, 103–110 (2009). 12. Rowe JA, Claessens A, Corrigan RA, Arman

M. Adhesion of Plasmodium falciparuminfected erythrocytes to human cells: molecular mechanisms and therapeutic implications. Expert Rev. Mol. Med. 11, e16 (2009). 13. Kyriacou HM, Stone GN, Challis RJ et al.

Differential var gene transcription in Plasmodium falciparum isolates from patients

7

Grau & Craig

PfEMP1-DBL1alpha amino acid motifs in severe disease states of Plasmodium falciparum malaria. Proc. Natl Acad. Sci. USA 104, 15835–15840 (2007). 15. Bull PC, Buckee CO, Kyes S et al.

Plasmodium falciparum antigenic variation. Mapping mosaic var gene sequences onto a network of shared, highly polymorphic sequence blocks. Mol. Microbiol. 68, 1519–1534 (2008). 16. Magistrado PA, Staalsoe T, Theander TG,

Hviid L, Jensen AT. CD36 selection of 3D7 Plasmodium falciparum associated with severe childhood malaria results in reduced VAR4 expression. Malar. J. 7, 204 (2008). 17. Warimwe GM, Keane TM, Fegan G et al.

Plasmodium falciparum var gene expression is modified by host immunity. Proc. Natl Acad. Sci. USA 106(51), 21801–21806 (2009). 18. Staalsoe T, Nielsen MA, Vestergaard LS,

Jensen AT, Theander TG, Hviid L. In vitro selection of Plasmodium falciparum 3D7 for expression of variant surface antigens associated with severe malaria in African children. Parasite Immunol. 25, 421–427 (2003). 19. Jensen AT, Magistrado P, Sharp S et al.

26. Dondorp AM, Nyanoti M, Kager PA,

Mithwani S, Vreeken J, Marsh K. The role of reduced red cell deformability in the pathogenesis of severe falciparum malaria and its restoration by blood transfusion. Trans. R. Soc. Trop. Med. Hyg. 96, 282–286 (2002). 27. Dondorp AM, Ince C, Charunwatthana P

et al. Direct in vivo assessment of microcirculatory dysfunction in severe falciparum malaria. J. Infect. Dis. 197, 79–84 (2008). 28. Viebig NK, Wulbrand U, Forster R, Andrews

KT, Lanzer M, Knolle PA. Direct activation of human endothelial cells by Plasmodium falciparum-infected erythrocytes. Infect. Immun. 73, 3271–3277 (2005). 29. Tripathi AK, Sullivan DJ, Stins MF.

Plasmodium falciparum-infected erythrocytes increase intercellular adhesion molecule 1 expression on brain endothelium through NF‑kB. Infect. Immun. 74, 3262–3270 (2006).

30. Chakravorty SJ, Carret C, Nash GB, Ivens A,

Szestak T, Craig AG. Altered phenotype and gene transcription in endothelial cells, induced by Plasmodium falciparum-infected red blood cells: pathogenic or protective? Int. J. Parasitol. 37, 975–987 (2007).

ho

Plasmodium falciparum associated with severe childhood malaria preferentially expresses PfEMP1 encoded by group A var genes. J. Exp. Med. 199, 1179–1190 (2004).

et al. Prognostic significance of reduced red blood cell deformability in severe falciparum malaria. Am. J. Trop. Med. Hyg. 57, 507–511 (1997).

20. Carlson J, Helmby H, Hill AV, Brewster D,

31. Tripathi AK, Sha W, Shulaev V, Stins MF,

Sullivan DJ Jr. Plasmodium falciparuminfected erythrocytes induce NF-kappaB regulated inflammatory pathways in human cerebral endothelium. Blood 114, 4243–4252 (2009).

ut

Greenwood BM, Wahlgren M. Human cerebral malaria: association with erythrocyte rosetting and lack of anti-rosetting antibodies. Lancet 336, 1457–1460 (1990). 21. Rowe A, Obeiro J, Newbold CI, Marsh K.

A

Plasmodium falciparum rosetting is associated with malaria severity in Kenya. Infect. Immun. 63, 2323–2326 (1995). 22. Pain A, Ferguson DJ, Kai O et al. Platelet-

mediated clumping of Plasmodium falciparum-infected erythrocytes is a common adhesive phenotype and is associated with severe malaria. Proc. Natl Acad. Sci. USA 98, 1805–1810 (2001). 23. Arman M, Raza A, Tempest LJ et al.

Platelet-mediated clumping of Plasmodium falciparum infected erythrocytes is associated with high parasitemia but not severe clinical manifestations of malaria in African children. Am. J. Trop. Med. Hyg. 77, 943–946 (2007). 24. Ochola LB, Siddondo BR, Ocholla H et al.

Specific receptor usage in Plasmodium falciparum cytoadherence is associated with disease outcome. PLoS ONE 6, e14741 (2011).

8

36. Brown H, Hien TT, Day N et al. Evidence of

blood–brain barrier dysfunction in human cerebral malaria. Neuropathol. Appl. Neurobiol. 25, 331–340 (1999). 37. Susomboon P, Maneerat Y, Dekumyoy P et al.

Down-regulation of tight junction mRNAs in human endothelial cells co-cultured with Plasmodium falciparum-infected erythrocytes. Parasitol. Int. 55, 107–112 (2006). 38. Toure FS, Ouwe-Missi-Oukem-Boyer O,

Bisvigou U et al. Apoptosis: a potential triggering mechanism of neurological manifestation in Plasmodium falciparum malaria. Parasite Immunol. 30, 47–51 (2008). 39. Wassmer SC, Combes V, Candal FJ,

Juhan-Vague I, Grau GE. Platelets potentiate brain endothelial alterations induced by Plasmodium falciparum. Infect. Immun. 74, 645–653 (2006).

of

14. Normark J, Nilsson D, Ribacke U et al.

25. Dondorp AM, Angus BJ, Hardeman MR

40. Pankoui Mfonkeu JB, Gouado I, Fotso Kuaté

H et al. Elevated cell-specific microparticles are a biological marker for cerebral dysfunctions in human severe malaria. PLoS ONE 5, e13415 (2010).

ro

with cerebral malaria compared with hyperparasitaemia. Mol. Biochem. Parasitol. 150, 211–218 (2006).

rP

Review

32. Medana IM, Turner GD. Plasmodium

falciparum and the blood–brain barrier – contacts and consequences. J. Infect. Dis. 195, 921–923 (2007).

33. Tripathi AK, Sullivan DJ, Stins MF.

Plasmodium falciparum-infected erythrocytes decrease the integrity of human blood–brain barrier endothelial cell monolayers. J. Infect. Dis. 195, 942–950 (2007). 34. Faille D, Combes V, Mitchell AJ et al. Platelet

microparticles: a new player in malaria parasite cytoadherence to human brain endothelium. FASEB J. 23, 3449–3458 (2009). 35. 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. 6, e1001021 (2010).

Future Microbiol. (2012) 7(2)

41. Nantakomol D, Dondorp AM, Krudsood S

et al. Circulating red cell-derived microparticles in human malaria. J. Infect. Dis. 203, 700–706 (2011).

42. Yipp BG, Robbins SM, Resek ME, Baruch

DI, Looareesuwan S, Ho M. Src-family kinase signaling modulates the adhesion of Plasmodium falciparum on human microvascular endothelium under flow. Blood 101, 2850–2857 (2003). 43. Ho M, Hoang HL, Lee KM et al.

Ectophosphorylation of CD36 regulates cytoadherence of Plasmodium falciparum to microvascular endothelium under flow conditions. Infect. Immun. 73, 8179–8187 (2005). 44. Dondorp AM, Silamut K, Charunwatthana P

et al. Levamisole inhibits sequestration of infected red blood cells in patients with falciparum malaria. J. Infect. Dis. 196, 460–466 (2007). 45. Ho M, Davis TM, Silamut K, Bunnag D,

White NJ. Rosette formation of Plasmodium falciparum-infected erythrocytes from patients with acute malaria. Infect. Immun. 59, 2135–2139 (1991). 46. Wassmer SC, Lepolard C, Traore B, Pouvelle

B, Gysin J, Grau GE. Platelets reorient Plasmodium falciparum-infected erythrocyte cytoadhesion to activated endothelial cells. J. Infect. Dis. 189, 180–189 (2004). 47. Bridges DJ, Bunn J, van Mourik JA et al.

Rapid activation of endothelial cells enables Plasmodium falciparum adhesion to platelet-decorated von Willebrand factor strings. Blood 115, 1472–1474 (2010).

future science group

Cerebral malaria pathogenesis: revisiting parasite & host contributions

48. Turner GD, Morrison H, Jones M 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. 145, 1057–1069 (1994).

58. Coban C, Ishii KJ, Sullivan DJ, Kumar N.

Purified malaria pigment (hemozoin) enhances dendritic cell maturation and modulates the isotype of antibodies induced by a DNA vaccine. Infect. Immun. 70, 3939–3943 (2002).

Marsh K, Craig A. Plasmodium falciparum intercellular adhesion molecule-1-based cytoadherence-related signaling in human endothelial cells. J. Infect. Dis. 196, 321–327 (2007).

Garside P, Brewer JM. Suppression of adaptive immunity to heterologous antigens during Plasmodium infection through hemozoin-induced failure of dendritic cell function. J. Biol. 5, 5 (2006). Inhibition of erythropoiesis in malaria anemia: role of hemozoin and hemozoingenerated 4-hydroxynonenal. Blood 116, 4328–4337 (2010).

host cells by a glycosylphosphatidylinositol toxin of malaria parasites. J. Exp. Med. 177, 145–153 (1993).

52. Tachado SD, Gerold P, McConville MJ et al.

53. Wichmann D, Schwarz RT, Ruppert V

54. Wennicke K, Debierre-Grockiego F,

A

Wichmann D et al. Glycosylphosphatidylinositol-induced cardiac myocyte death might contribute to the fatal outcome of Plasmodium falciparum malaria. Apoptosis 13, 857–866 (2008). 55. Shio MT, Kassa FA, Bellemare MJ, Olivier

M. Innate inflammatory response to the malarial pigment hemozoin. Microbes Infect. 12, 889–899 (2010). 56. Nguyen PH, Day N, Pram TD, Ferguson DJ,

White NJ. Intraleucocytic malaria pigment and prognosis in severe malaria. Trans. R. Soc. Trop. Med. Hyg. 89, 200–204 (1995). 57. Amodu OK, Adeyemo AA, Olumese PE,

Gbadegesin RA. Intraleucocytic malaria pigment and clinical severity of malaria in children. Trans. R. Soc. Trop. Med. Hyg. 92, 54–56 (1998).

future science group

71. MacDonald SM, Bhisutthibhan J, Shapiro TA

et al. Immune mimicry in malaria: Plasmodium falciparum secretes a functional histamine-releasing factor homolog in vitro and in vivo. Proc. Natl Acad. Sci. USA 98, 10829–10832 (2001). 72. Maegraith B. Pathological processes in

malaria. Trans. R. Soc. Trop. Med. Hyg. 41, 687–704 (1948).

of

62. Prato M, Giribaldi G, Polimeni M, Gallo V,

Arese P. Phagocytosis of hemozoin enhances matrix metalloproteinase-9 activity and TNF-alpha production in human monocytes: role of matrix metalloproteinases in the pathogenesis of falciparum malaria. J. Immunol. 175, 6436–6442 (2005).

63. Prato M, D’Alessandro S, Van den Steen PE

et al. Natural haemozoin modulates matrix metalloproteinases and induces morphological changes in human microvascular endothelium. Cell. Microbiol. 13, 1275–1285 (2011).

64. Conroy AL, Phiri H, Hawkes M et al.

ut

et al. Plasmodium falciparum glycosylphosphatidylinositol induces limited apoptosis in liver and spleen mouse tissue. Apoptosis 12, 1037–1041 (2007).

Voorberg J, Eikenboom J. Functional architecture of Weibel-Palade bodies. Blood 117, 5033–5043 (2011).

73. Maegraith B. The physiological approach to

the problems of malaria. Br. Med. Bull. 8, 28–32 (1951).

74. Coltel N, Combes V, Hunt NH, Grau GE.

ro

Arese P, Schwarzer E. Host fibrinogen stably bound to hemozoin rapidly activates monocytes via TLR-4 and CD11b/ CD18-integrin: a new paradigm of hemozoin action. Blood 117, 5674–5682 (2011).

ho

Glycosylphosphatidylinositol toxin of Plasmodium induces nitric oxide synthase expression in macrophages and vascular endothelial cells by a protein tyrosine kinase-dependent and protein kinase C-dependent signaling pathway. J. Immunol. 156, 1897–1907 (1996).

70. Valentijn KM, Sadler JE, Valentijn JA,

61. Barrera V, Skorokhod OA, Baci D, Gremo G,

rP

RT, McConville MJ, Tachado SD. Glycosylphosphatidylinositol toxin of Plasmodium up-regulates intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and E-selectin expression in vascular endothelial cells and increases leukocyte and parasite cytoadherence via tyrosine kinase-dependent signal transduction. J. Immunol. 156, 1886–1896 (1996).

et al. Severe malaria is associated with a deficiency of von Willebrand factor cleaving protease, ADAMTS13. Thromb. Haemost. 103, 181–187 (2010).

60. Skorokhod OA, Caione L, Marrocco T et al.

50. Schofield L, Hackett F. Signal transduction in

51. Schofield L, Novakovic S, Gerold P, Schwarz

69. Lowenberg EC, Charunwatthana P, Cohen S

59. Millington OR, Di Lorenzo C, Phillips RS,

49. Jenkins N, Wu Y, Chakravorty S, Kai O,

Review

Endothelium-based biomarkers are associated with cerebral malaria in Malawian children: a retrospective case–control study. PLoS ONE 5, e15291 (2010).

65. Hollestelle MJ, Donkor C, Mantey EA et al.

von Willebrand factor propeptide in malaria: evidence of acute endothelial cell activation. Br. J. Haematol. 133, 562–569 (2006).

66. de Mast Q, Groot E, Lenting PJ et al.

Thrombocytopenia and release of activated von Willebrand Factor during early Plasmodium falciparum malaria. J. Infect. Dis. 196, 622–628 (2007). 67. de Mast Q, Groot E, Asih PB et al.

ADAMTS13 deficiency with elevated levels of ultra-large and active von Willebrand factor in P. falciparum and P. vivax malaria. Am. J. Trop. Med. Hyg. 80, 492–498 (2009). 68. Larkin D, de Laat B, Jenkins PV et al. Severe

Plasmodium falciparum malaria is associated with circulating ultra-large von Willebrand multimers and ADAMTS13 inhibition. PLoS Pathog. 5, e1000349 (2009).

www.futuremedicine.com

Cerebral malaria – a neurovascular pathology with many riddles still to be solved. Curr. Neurovasc. Res. 1, 91–110 (2004).

75. Hunt NH, Grau GE. Cytokines: accelerators

and brakes in the pathogenesis of cerebral malaria. Trends Immunol. 24, 491–499 (2003). 76. Schofield L, Grau GE. Immunological

processes in malaria pathogenesis. Trends Immunol. 5, 722–735 (2005). 77. Grau GE, Taylor TE, Molyneux ME et al.

Tumor necrosis factor and disease severity in children with falciparum malaria. N. Engl. J. Med. 320, 1586–1591 (1989). 78. Kwiatkowski D, Hill AV, Sambou I et al.

TNF concentration in fatal cerebral, non-fatal cerebral, and uncomplicated Plasmodium falciparum malaria. Lancet 336, 1201–1204 (1990). 79. Kern P, Hemmer CJ, Van Damme J, Gruss

HJ, Dietrich M. Elevated tumor necrosis factor alpha and interleukin-6 serum levels as markers for complicated Plasmodium falciparum malaria. Am. J. Med. 87, 139–143 (1989). 80. Riley EM, Couper KN, Helmby H et al.

Neuropathogenesis of human and murine malaria. Trends Parasitol. 26, 277–278 (2010). 81. Francischetti IM, Seydel KB, Monteiro RQ

et al. Plasmodium falciparum-infected erythrocytes induce tissue factor expression in endothelial cells and support the assembly of multimolecular coagulation complexes. J. Thromb. Haemost. 5, 155–165 (2007). 82. Lopez JA. Malignant malaria and

microangiopathies: merging mechanisms. Blood 115, 1317–1318 (2010).

9

Grau & Craig

84. Lovegrove FE, Tangpukdee N, Opoka RO

et al. Serum angiopoietin-1 and -2 levels discriminate cerebral malaria from uncomplicated malaria and predict clinical outcome in African children. PLoS ONE 4, e4912 (2009). 85. Combes V, De Souza JB, Renia L, Hunt NH,

Grau GE. Cerebral malaria: which parasite? which model? Drug Discov. Today: Disease Models 2, 141–148 (2005). 86. Grau GE, Fajardo LF, Piguet PF, Allet B,

Lambert PH, Vassalli P. Tumor necrosis factor (cachectin) as an essential mediator in murine cerebral malaria. Science 237, 1210–1212 (1987). 87. Grau GE, Heremans H, Piguet PF et al.

Monoclonal antibody against interferon gamma can prevent experimental cerebral malaria and its associated overproduction of tumor necrosis factor. Proc. Natl Acad. Sci. USA 86, 5572–5574 (1989). 88. Engwerda CR, Mynott TL, Sawhney S, De

Souza JB, Bickle QD, Kaye PM. Locally up-regulated lymphotoxin alpha, not systemic tumor necrosis factor alpha, is the principle mediator of murine cerebral malaria. J. Exp. Med. 195, 1371–1377 (2002). 89. Schmidt KE, Schumak B, Specht S, Dubben

96. Hunt NH, Stocker R. Heme moves to center

stage in cerebral malaria. Nat. Med. 13, 667–669 (2007). 97. van der Heyde HC, Nolan J, Combes V,

Gramaglia I, Grau GE. A unified hypothesis for the genesis of cerebral malaria: sequestration, inflammation and hemostasis leading to microcirculatory dysfunction. Trends Parasitol. 22, 503–508 (2006). 98. Grau GE, Mackenzie CD, Carr RA et al.

Platelet accumulation in brain microvessels in fatal pediatric cerebral malaria. J. Infect. Dis. 187, 461–466 (2003). 99. Chirenda J, Murugasampillay S. Malaria and

HIV co-infection: available evidence, gaps and possible interventions. Cent. Afr. J. Med. 49, 66–71 (2003). 100. Chandramohan D, Greenwood BM. Is there

an interaction between human immunodeficiency virus and Plasmodium falciparum? Int. J. Epidemiol. 27, 296–301 (1998). 101. Bejon P, Berkley JA, Mwangi T et al.

Defining childhood severe falciparum malaria for intervention studies. PLoS Med. 4, e251 (2007).

102. Rogerson SJ, Wijesinghe RS, Meshnick SR.

Host immunity as a determinant of treatment outcome in Plasmodium falciparum malaria. Lancet Infect. Dis. 10, 51–59 (2010).

ho

B, Limmer A, Hoerauf A. Induction of pro-inflammatory mediators in Plasmodium berghei infected BALB/c mice breaks blood– brain-barrier and leads to cerebral malaria in an IL-12 dependent manner. Microbes Infect. 13(10), 828–836 (2011).

cerebral malaria. Exp. Biol. Med. (Maywood) 231, 1176–1181 (2006).

103. Grau GE, Piguet PF, Engers HD, Louis JA,

Vassalli P, Lambert PH. L3T4 T lymphocytes play a major role in the pathogenesis of murine cerebral malaria. J. Immunol. 137, 2348–2354 (1986). +

104. Amante FH, Haque A, Stanley AC et al.

ut

90. Campanella GS, Tager AM, El Khoury JK

A

et al. Chemokine receptor CXCR3 and its ligands CXCL9 and CXCL10 are required for the development of murine cerebral malaria. Proc. Natl Acad. Sci. USA 105, 4814–4819 (2008). 91. Mitchell AJ, Hansen AM, Hee L et al. Early

cytokine production is associated with protection from murine cerebral malaria. Infect. Immun. 73, 5645–5653 (2005). 92. Beghdadi W, Porcherie A, Schneider BS et al.

[Role of histamine and histamine receptors in the pathogenesis of malaria]. Med. Sci. (Paris) 25, 377–381 (2009). 93. Beghdadi W, Porcherie A, Schneider BS et al.

Histamine H(3) receptor-mediated signaling protects mice from cerebral malaria. PLoS ONE 4, e6004 (2009). 94. Patel SN, Berghout J, Lovegrove FE et al. C5

deficiency and C5a or C5aR blockade protects against cerebral malaria. J. Exp. Med. 205, 1133–1143 (2008). 95. Machado FS, Desruisseaux MS, Nagajyothi

et al. Endothelin in a murine model of

10

109. Nie CQ, Bernard NJ, Schofield L, Hansen

DS. CD4+ CD25+ regulatory T cells suppress CD4+ T-cell function and inhibit the development of Plasmodium berghei-specific TH1 responses involved in cerebral malaria pathogenesis. Infect. Immun. 75, 2275–2282 (2007). 110. Haque A, Best SE, Amante FH et al. CD4+

natural regulatory T cells prevent experimental cerebral malaria via CTLA-4 when expanded in vivo. PLoS Pathog. 6, e1001221 (2010). 111. Torcia MG, Santarlasci V, Cosmi L et al.

Functional deficit of T regulatory cells in Fulani, an ethnic group with low susceptibility to Plasmodium falciparum malaria. Proc. Natl Acad. Sci. USA 105, 646–651 (2008). 112. Minigo G, Woodberry T, Piera KA et al.

Parasite-dependent expansion of TNF receptor II-positive regulatory T cells with enhanced suppressive activity in adults with severe malaria. PLoS Pathog. 5, e1000402 (2009).

of

Combinations of host biomarkers predict mortality among Ugandan children with severe malaria: a retrospective case–control study. PLoS ONE 6, e17440 (2011).

ro

83. Erdman LK, Dhabangi A, Musoke C et al.

rP

Review

Immune-mediated mechanisms of parasite tissue sequestration during experimental cerebral malaria. J. Immunol. 185, 3632–3642 (2010).

105. Yanez 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. 157, 1620–1624 (1996). 106. Belnoue E, Kayibanda M, Vigario AM et al.

On the pathogenic role of brain-sequestered alphabeta CD8+ T cells in experimental cerebral malaria. J. Immunol. 169, 6369–6375 (2002). 107. Hunt NH, Grau GE, Engwerda C et al.

Murine cerebral malaria: the whole story. Trends Parasitol. 26, 272–274 (2010). 108. Amante FH, Stanley AC, Randall LM et al. A

role for natural regulatory T cells in the pathogenesis of experimental cerebral malaria. Am. J. Pathol. 171, 548–559 (2007).

Future Microbiol. (2012) 7(2)

113. White VA, Lewallen S, Beare NA, Molyneux

ME, Taylor TE. Retinal pathology of pediatric cerebral malaria in Malawi. PLoS ONE 4, e4317 (2009).

114. Grau GE, Tacchini-Cottier F, Vesin C et al.

TNF-induced microvascular pathology: active role for platelets and importance of the LFA-1/ ICAM-1 interaction. Eur. Cytokine Netw. 4, 415–419 (1993). 115. Grau GE, Lou J. TNF in vascular pathology:

the importance of platelet-endothelium interactions. Res. Immunol. 144, 355–363 (1993). 116. von Zur Muhlen C, Sibson NR, Peter K et al.

A contrast agent recognizing activated platelets reveals murine cerebral malaria pathology undetectable by conventional MRI. J. Clin. Invest. 118, 1198–1207 (2008). 117. Lou J, Donati YR, Juillard P et al. Platelets

play an important role in TNF-induced microvascular endothelial cell pathology. Am. J. Pathol. 151, 1397–1405 (1997). 118. Wassmer SC, de Souza JB, Frere C, Candal

FJ, Juhan-Vague I, Grau GE. TGF-beta1 released from activated platelets can induce TNF-stimulated human brain endothelium apoptosis: a new mechanism for microvascular lesion during cerebral malaria. J. Immunol. 176, 1180–1184 (2006). 119. Barbier M, Faille D, Loriod B et al. Platelets

alter gene expression profile in human brain endothelial cells in an in vitro model of cerebral malaria. PLoS ONE 6, e19651 (2011). 120. 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. 91, 140–151 (2010).

future science group

Cerebral malaria pathogenesis: revisiting parasite & host contributions

cerebral malaria: a randomized trial. Clin. Infect. Dis. 53, 349–355 (2011).

121. Combes V, Lou J, Grau GE. Microvascular

endothelium and cerebral malaria. In: Endothelium Biomedicine: A Comprehensve Reference. Aird W (Ed.). Cambridge University Press, NY, USA, 1303–1310 (2007). 122. El-Assaad F, Hempel C, Combes V et al.

Differential microRNA expression in experimental cerebral and noncerebral malaria. Infect. Immun. 79, 2379–2384 (2011).

Angiopoietin-2 is associated with decreased endothelial nitric oxide and poor clinical outcome in severe falciparum malaria. Proc. Natl Acad. Sci. USA 105, 17097–17102 (2008). 136. Jain V, Nagpal AC, Joel PK et al. Burden of

Recovery of endothelial function in severe falciparum malaria: relationship with improvement in plasma l-arginine and blood lactate concentrations. J. Infect. Dis. 198, 602–608 (2008).

139. Chakravorty SJ, Hughes KR, Craig AG. Host

response to cytoadherence in Plasmodium falciparum. Biochem. Soc. Trans. 36, 221–228 (2008).

ho

et al. Magnetic resonance imaging of the brain in patients with cerebral malaria. Clin. Infect. Dis. 21, 300–309 (1995).

140. Wassmer SC, Moxon CA, Taylor T, Grau GE,

128. Saavedra-Lozano J, Booth TN, Weprin BE,

ut

Ramilo O. Isolated cerebellar edema and obstructive hydrocephalus in a child with cerebral malaria. Pediatr. Infect. Dis. J. 20, 908–911 (2001).

129. Penet MF, Viola A, Confort-Gouny S et al.

A

Imaging experimental cerebral malaria in vivo: significant role of ischemic brain edema. J. Neurosci. 25, 7352–7358 (2005). 130. Saggu R, Faille D, Grau GE, Cozzone PJ,

Viola A. In the eye of experimental cerebral malaria. Am. J. Pathol. 179(3), 1104–1109 (2011). 131. Ampawong S, Combes V, Hunt NH et al.

Quantitation of brain edema and localisation of aquaporin 4 expression in relation to susceptibility to experimental cerebral malaria. Int. J. Clin. Exp. Pathol. 4, 566–574 (2011). 132. Medana IM, Day NP, Sachanonta N et al.

Coma in fatal adult human malaria is not caused by cerebral oedema. Malar. J. 10, 267 (2011). 133. Mohanty S, Mishra SK, Patnaik R et al. Brain

swelling and mannitol therapy in adult

future science group

Glial activation and matrix metalloproteinase release in cerebral malaria. J. Neurovirol. 13, 2–10 (2007). 149. Miu J, Mitchell AJ, Muller M et al.

Chemokine gene expression during fatal murine cerebral malaria and protection due to CXCR3 deficiency. J. Immunol. 180, 1217–1230 (2008).

of

138. Yeo TW, Lampah DA, Gitawati R et al.

rP

127. Looareesuwan S, Wilairatana P, Krishna S

148. Szklarczyk A, Stins M, Milward EA et al.

150. Medana IM, Mai NT, Day NP et al. Cellular

ro

Impaired nitric oxide bioavailability and l-arginine reversible endothelial dysfunction in adults with falciparum malaria. J. Exp. Med. 204, 2693–2704 (2007).

126. Lou J, Dayer JM, Grau GE, Burger D. Direct

cell/cell contact with stimulated T lymphocytes induces the expression of cell adhesion molecules and cytokines by human brain microvascular endothelial cells. Eur. J. Immunol. 26, 3107–3113 (1996).

Soluble factors from Plasmodium falciparuminfected erythrocytes induce apoptosis in human brain vascular endothelial and neuroglia cells. Mol. Biochem. Parasitol. 162, 172–176 (2008).

137. Yeo TW, Lampah DA, Gitawati R et al.

125. Medana IM, Turner GDH. Human cerebral

malaria and the blood–brain barrier. Int. J. Parasitol. 36, 555–568 (2006).

147. Wilson NO, Huang MB, Anderson W et al.

cerebral malaria in central India (2004–2007). Am. J. Trop. Med. Hyg. 79, 636–642 (2008).

124. Turner GD, Ly VC, Nguyen TH et al.

Systemic endothelial activation occurs in both mild and severe malaria. Correlating dermal microvascular endothelial cell phenotype and soluble cell adhesion molecules with disease severity. Am. J. Pathol. 152, 1477–1487 (1998).

R, Schluesener H. Macrophages/microglial cells in patients with cerebral malaria. Eur. Cytokine Netw. 13, 173–185 (2002).

135. Yeo TW, Lampah DA, Gitawati R et al.

123. Porta J, Carota A, Pizzolato GP et al.

Immunopathological changes in human cerebral malaria. Clin. Neuropathol. 12, 142–146 (1993).

146. Deininger MH, Kremsner PG, Meyermann

134. Furuta T, Kimura M, Watanabe N. Elevated

levels of vascular endothelial growth factor (VEGF) and soluble vascular endothelial growth factor receptor (VEGFR)-2 in human malaria. Am. J. Trop. Med. Hyg. 82, 136–139 (2010).

Review

Molyneux ME, Craig AG. Vascular endothelial cells cultured from patients with cerebral or uncomplicated malaria exhibit differential reactivity to TNF. Cell. Microbiol. 13, 198–209 (2011).

141. Lou J, Gasche Y, Zheng L et al. Differential

reactivity of brain microvascular endothelial cells to TNF reflects the genetic susceptibility to cerebral malaria. Eur. J. Immunol. 28, 3989–4000 (1998).

142. Combes V, Taylor TE, Juhan-Vague I et al.

Circulating endothelial microparticles in malawian children with severe falciparum malaria complicated with coma. JAMA 291, 2542–2544 (2004). 143. Coltel N, Combes V, Wassmer SC, Chimini

G, Grau GE. Cell vesiculation and immunopathology: implications in cerebral malaria. Microbes Infect. 8, 2305–2316 (2006). 144. Combes V, Coltel N, Faille D, Wassmer SC,

Grau GE. Cerebral malaria: role of microparticles and platelets in alterations of the blood–brain barrier. Int. J. Parasitol. 36, 541–546 (2006). 145. Wassmer et al. XXXXXX Drug Discov. Today

(2012) (In Press).

www.futuremedicine.com

stress and injury responses in the brains of adult Vietnamese patients with fatal Plasmodium falciparum malaria. Neuropathol. Appl. Neurobiol. 27, 421–433 (2001).

151. Medana IM, Idro R, Newton CR. Axonal and

astrocyte injury markers in the cerebrospinal fluid of Kenyan children with severe malaria. J. Neurol. Sci. 258, 93–98 (2007).

152. Medana IM, Day NP, Hien TT et al.

Cerebral calpain in fatal falciparum malaria. Neuropathol. Appl. Neurobiol. 33, 179–192 (2007). 153. 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 19, 135–151 (1997). 154. Hunt NH, Golenser J, Chan-Ling T et al.

Immunopathogenesis of cerebral malaria. Int. J. Parasitol. 36, 569–582 (2006). 155. DellaValle B, Hempel C, Kurtzhals JA,

Penkowa M. In vivo expression of neuroglobin in reactive astrocytes during neuropathology in murine models of traumatic brain injury, cerebral malaria, and autoimmune encephalitis. Glia 58, 1220–1227 (2010). 156. Carter JA, Mung’ala-Odera V, Neville BG

et al. Persistent neurocognitive impairments associated with severe falciparum malaria in Kenyan children. J. Neurol. Neurosurg. Psychiatry 76, 476–481 (2005). 157. Carter JA, Ross AJ, Neville BG et al.

Developmental impairments following severe falciparum malaria in children. Trop. Med. Int. Health 10, 3–10 (2005). 158. Bangirana P, Idro R, John CC, Boivin

MJ. Rehabilitation for cognitive impairments after cerebral malaria in African children: strategies and limitations. Trop. Med. Int. Health 11, 1341–1349 (2006).

11

Review

Grau & Craig

159. Bangirana P, Allebeck P, Boivin MJ et al.

Cognition, behaviour and academic skills after cognitive rehabilitation in Ugandan children surviving severe malaria: a randomised trial. BMC Neurol. 11, 96 (2011). 160. Idro R, Kakooza-Mwesige A, Balyejjussa S

161. Lacerda-Queiroz N, Rodrigues DH, Vilela

MC et al. Inflammatory changes in the central nervous system are associated with behavioral impairment in Plasmodium berghei

(strain ANKA)-infected mice. Exp. Parasitol. 125, 271–278 (2010). 162. Reis PA, Comim CM, Hermani F et al.

Cognitive dysfunction is sustained after rescue therapy in experimental cerebral malaria, and is reduced by additive antioxidant therapy. PLoS Pathog. 6, e1000963 (2010).

A

ut

ho

rP

ro

of

et al. Severe neurological sequelae and

behaviour problems after cerebral malaria in Ugandan children. BMC Res. Notes 3, 104 (2010).

12

Future Microbiol. (2012) 7(2)

future science group