Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Radiology ; 290(2): 359-367, 2019 02.
Article in English | MEDLINE | ID: mdl-30615566

ABSTRACT

Purpose To investigate the association of inflammation and brain edema in a cerebral malaria (CM) mouse model with a combination of bis-5-hydroxy-tryptamide-diethylenetriaminepentaacetate gadolinium, referred to as MPO-Gd, and cross-linked iron oxide nanoparticle (CLIO-NP) imaging. Materials and Methods Female wild-type (n = 23) and myeloperoxidase (MPO) knock-out (n = 5) mice were infected with the Plasmodium berghei ANKA strain from May 2016 to July 2018. Seven healthy mice served as control animals. At a Rapid Murine Coma and Behavioral Scale (RMCBS) score of less than 15, mice underwent MRI at 9.4 T and received gadodiamide, MPO-Gd, or CLIO-NPs. T1-weighted MRI was used to assess MPO activity, and T2*-weighted MRI was used to track CLIO-NPs. Immunofluorescent staining and flow cytometric analyses characterized CLIO-NPs, MPO, endothelial cells, and leukocytes. An unpaired, two-tailed Student t test was used to compare groups; Spearman correlation analysis was used to determine the relationship of imaging parameters to clinical severity. Results MPO-Gd enhancement occurred in inflammatory CM hotspots (olfactory bulb > rostral migratory stream > brainstem > cortex, P < .05 for all regions compared with control mice; mean olfactory bulb signal intensity ratio: 1.40 ± 0.07 vs 0.96 ± 0.01, P < .01). The enhancement was reduced in MPO knockout mice (mean signal intensity ratio at 60 minutes: 1.13 ± 0.04 vs 1.40 ± 0.07 in CM, P < .05). Blood-brain barrier compromise was suggested by parenchymal gadolinium enhancement, leukocyte recruitment, and endothelial activation. CLIO-NPs accumulated mainly intravascularly and at the vascular endothelium. CLIO-NPs were also found in the choroid plexus, indicating inflammation of the ventricular system. Blood-cerebrospinal fluid barrier breakdown showed correlation with brain swelling (r2: 0.55, P < .01) and RMCBS score (r2: 0.75, P < .001). Conclusion Iron oxide nanoparticle imaging showed strong inflammatory involvement of the microvasculature in a murine model of cerebral malaria. Furthermore, bis-5-hydroxy-tryptamide-diethylenetriaminepentaacetate gadolinium imaging depicted parenchymal and intraventricular inflammation. This combined molecular imaging approach links vascular inflammation to breakdown of the blood-brain barrier and blood-cerebrospinal fluid barrier that correlate with global brain edema and disease severity. © RSNA, 2018 Online supplemental material is available for this article. See also the editorial by Kiessling in this issue.


Subject(s)
Brain Edema , Encephalitis , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles/chemistry , Malaria, Cerebral , Peroxidase/metabolism , Animals , Brain/diagnostic imaging , Brain/enzymology , Brain/pathology , Brain Edema/diagnostic imaging , Brain Edema/enzymology , Brain Edema/parasitology , Brain Edema/pathology , Disease Models, Animal , Encephalitis/diagnostic imaging , Encephalitis/enzymology , Encephalitis/parasitology , Encephalitis/pathology , Female , Malaria, Cerebral/complications , Malaria, Cerebral/diagnostic imaging , Malaria, Cerebral/enzymology , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
Nat Med ; 13(6): 703-10, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17496899

ABSTRACT

Cerebral malaria claims more than 1 million lives per year. We report that heme oxygenase-1 (HO-1, encoded by Hmox1) prevents the development of experimental cerebral malaria (ECM). BALB/c mice infected with Plasmodium berghei ANKA upregulated HO-1 expression and activity and did not develop ECM. Deletion of Hmox1 and inhibition of HO activity increased ECM incidence to 83% and 78%, respectively. HO-1 upregulation was lower in infected C57BL/6 compared to BALB/c mice, and all infected C57BL/6 mice developed ECM (100% incidence). Pharmacological induction of HO-1 and exposure to the end-product of HO-1 activity, carbon monoxide (CO), reduced ECM incidence in C57BL/6 mice to 10% and 0%, respectively. Whereas neither HO-1 nor CO affected parasitemia, both prevented blood-brain barrier (BBB) disruption, brain microvasculature congestion and neuroinflammation, including CD8(+) T-cell brain sequestration. These effects were mediated by the binding of CO to hemoglobin, preventing hemoglobin oxidation and the generation of free heme, a molecule that triggers ECM pathogenesis.


Subject(s)
Carbon Monoxide/physiology , Heme Oxygenase-1/physiology , Heme/metabolism , Malaria, Cerebral/enzymology , Animals , Disease Models, Animal , Heme Oxygenase-1/deficiency , Heme Oxygenase-1/genetics , Malaria, Cerebral/drug therapy , Malaria, Cerebral/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Plasmodium berghei
3.
Am J Pathol ; 178(1): 212-21, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21224058

ABSTRACT

Cerebral malaria is the most severe neurologic complication in children and young adults infected with Plasmodium falciparum. T-cell activation is required for development of Plasmodium berghei ANKA (PbA)-induced experimental cerebral malaria (CM). To characterize the T-cell activation pathway involved, the role of protein kinase C-theta (PKC-θ) in experimental CM development was examined. PKC-θ-deficient mice are resistant to CM development. In the absence of PKC-θ, no neurologic sign of CM developed after blood stage PbA infection. Resistance of PKC-θ-deficient mice correlated with unaltered cerebral microcirculation and absence of ischemia, as documented by magnetic resonance imaging and magnetic resonance angiography, whereas wild-type mice developed distinct microvascular pathology. Recruitment and activation of CD8(+) T cells, and ICAM-1 and CD69 expression were reduced in the brain of resistant mice; however, the pulmonary inflammation and edema associated with PbA infection were still present in the absence of functional PKC-θ. Resistant PKC-θ-deficient mice developed high parasitemia, and died at 3 weeks with severe anemia. Therefore, PKC-θ signaling is crucial for recruitment of CD8(+) T cells and development of brain microvascular pathology resulting in fatal experimental CM, and may represent a novel therapeutic target of CM.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Isoenzymes/metabolism , Malaria, Cerebral/enzymology , Malaria, Cerebral/immunology , Plasmodium berghei , Protein Kinase C/metabolism , Animals , Brain/blood supply , Brain/parasitology , Brain/pathology , Brain Ischemia/enzymology , Brain Ischemia/immunology , Brain Ischemia/pathology , Cell Movement , Disease Models, Animal , Isoenzymes/genetics , Magnetic Resonance Angiography , Magnetic Resonance Imaging , Malaria, Cerebral/pathology , Mice , Mice, Mutant Strains , Microcirculation , Microvessels/pathology , Parasitemia/enzymology , Parasitemia/immunology , Protein Kinase C/genetics , Protein Kinase C-theta
4.
Nat Commun ; 11(1): 4596, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32929083

ABSTRACT

Earlier studies indicate that either the canonical or non-canonical pathways of inflammasome activation have a limited role on malaria pathogenesis. Here, we report that caspase-8 is a central mediator of systemic inflammation, septic shock in the Plasmodium chabaudi-infected mice and the P. berghei-induced experimental cerebral malaria (ECM). Importantly, our results indicate that the combined deficiencies of caspases-8/1/11 or caspase-8/gasdermin-D (GSDM-D) renders mice impaired to produce both TNFα and IL-1ß and highly resistant to lethality in these models, disclosing a complementary, but independent role of caspase-8 and caspases-1/11/GSDM-D in the pathogenesis of malaria. Further, we find that monocytes from malaria patients express active caspases-1, -4 and -8 suggesting that these inflammatory caspases may also play a role in the pathogenesis of human disease.


Subject(s)
Caspase 8/metabolism , Inflammation/pathology , Malaria, Cerebral/enzymology , Animals , Brain/pathology , Caspase 1/metabolism , Dendritic Cells/metabolism , Enzyme Activation , Extracellular Matrix/metabolism , Gene Expression Regulation , Humans , Interferon-gamma/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides , Malaria, Cerebral/genetics , Mice, Inbred C57BL , Monocytes/metabolism , Plasmodium chabaudi/physiology , Spleen/metabolism , Toll-Like Receptors/metabolism
5.
Cell Rep ; 32(12): 108170, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32966787

ABSTRACT

The replication cycle and pathogenesis of the Plasmodium malarial parasite involves rapid expansion in red blood cells (RBCs), and variants of certain RBC-specific proteins protect against malaria in humans. In RBCs, bisphosphoglycerate mutase (BPGM) acts as a key allosteric regulator of hemoglobin/oxyhemoglobin. We demonstrate here that a loss-of-function mutation in the murine Bpgm (BpgmL166P) gene confers protection against both Plasmodium-induced cerebral malaria and blood-stage malaria. The malaria protection seen in BpgmL166P mutant mice is associated with reduced blood parasitemia levels, milder clinical symptoms, and increased survival. The protective effect of BpgmL166P involves a dual mechanism that enhances the host's stress erythroid response to Plasmodium-driven RBC loss and simultaneously alters the intracellular milieu of the RBCs, including increased oxyhemoglobin and reduced energy metabolism, reducing Plasmodium maturation, and replication. Overall, our study highlights the importance of BPGM as a regulator of hemoglobin/oxyhemoglobin in malaria pathogenesis and suggests a new potential malaria therapeutic target.


Subject(s)
Anemia/etiology , Anemia/prevention & control , Bisphosphoglycerate Mutase/deficiency , Malaria, Cerebral/enzymology , Malaria, Cerebral/prevention & control , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , Bisphosphoglycerate Mutase/chemistry , Bisphosphoglycerate Mutase/genetics , Bisphosphoglycerate Mutase/metabolism , Enzyme Stability , Erythrocytes/enzymology , Erythrocytes/parasitology , Erythropoiesis , Extracellular Matrix/metabolism , Female , HEK293 Cells , Humans , Malaria, Cerebral/complications , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation/genetics , Parasites/growth & development , Plasmodium/growth & development , Polycythemia
7.
J Mol Med (Berl) ; 86(10): 1097-111, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18641963

ABSTRACT

Malaria, the disease caused by Plasmodium infection, is endemic to poverty in so-called underdeveloped countries. Plasmodium falciparum, the main infectious Plasmodium species in sub-Saharan countries, can trigger the development of severe malaria, including cerebral malaria, a neurological syndrome that claims the lives of more than one million children (<5 years old) per year. Attempts to eradicate Plasmodium infection, and in particular its lethal outcomes, have so far been unsuccessful. Using well-established rodent models of malaria infection, we found that survival of a Plasmodium-infected host is strictly dependent on the host's ability to up-regulate the expression of heme oxygenase-1 (HO-1 encoded by the gene Hmox1). HO-1 is a stress-responsive enzyme that catabolizes free heme into biliverdin, via a reaction that releases Fe and generates the gas carbon monoxide (CO). Generation of CO through heme catabolism by HO-1 prevents the onset of cerebral malaria. The protective effect of CO is mediated via its binding to cell-free hemoglobin (Hb) released from infected red blood cells during the blood stage of Plasmodium infection. Binding of CO to cell-free Hb prevents heme release and thus generation of free heme, which we found to play a central role in the pathogenesis of cerebral malaria. We will address hereby how defense mechanisms that prevent the deleterious effects of free heme, including the expression of HO-1, impact on the pathologic outcome of Plasmodium infection and how these may be used therapeutically to suppress its lethal outcomes.


Subject(s)
Heme/physiology , Malaria/physiopathology , Animals , Carbon Monoxide/metabolism , Disease Models, Animal , Heme/metabolism , Heme Oxygenase-1/metabolism , Hemoglobins/metabolism , Humans , Malaria/enzymology , Malaria/metabolism , Malaria, Cerebral/enzymology , Malaria, Cerebral/metabolism , Malaria, Cerebral/physiopathology , Models, Biological
8.
EMBO Mol Med ; 11(8): e9903, 2019 08.
Article in English | MEDLINE | ID: mdl-31265218

ABSTRACT

Malaria is a highly inflammatory disease caused by Plasmodium infection of host erythrocytes. However, the parasite does not induce inflammatory cytokine responses in macrophages in vitro and the source of inflammation in patients remains unclear. Here, we identify oxidative stress, which is common in malaria, as an effective trigger of the inflammatory activation of macrophages. We observed that extracellular reactive oxygen species (ROS) produced by xanthine oxidase (XO), an enzyme upregulated during malaria, induce a strong inflammatory cytokine response in primary human monocyte-derived macrophages. In malaria patients, elevated plasma XO activity correlates with high levels of inflammatory cytokines and with the development of cerebral malaria. We found that incubation of macrophages with plasma from these patients can induce a XO-dependent inflammatory cytokine response, identifying a host factor as a trigger for inflammation in malaria. XO-produced ROS also increase the synthesis of pro-IL-1ß, while the parasite activates caspase-1, providing the two necessary signals for the activation of the NLRP3 inflammasome. We propose that XO-produced ROS are a key factor for the trigger of inflammation during malaria.


Subject(s)
Inflammation/enzymology , Macrophages/enzymology , Malaria, Cerebral/enzymology , Malaria, Falciparum/enzymology , Oxidative Stress , Plasmodium falciparum/pathogenicity , Reactive Oxygen Species/metabolism , Xanthine Oxidase/metabolism , Caspase 1/metabolism , Cells, Cultured , Cytokines/metabolism , Host-Pathogen Interactions , Humans , Inflammation/blood , Inflammation/parasitology , Inflammation Mediators/metabolism , Macrophage Activation , Macrophages/parasitology , Malaria, Cerebral/blood , Malaria, Cerebral/parasitology , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction
9.
Genes (Basel) ; 10(6)2019 06 21.
Article in English | MEDLINE | ID: mdl-31234346

ABSTRACT

Malaria is one of the three major global health threats. Drug development for malaria, especially for its most dangerous form caused by Plasmodium falciparum, remains an urgent task due to the emerging drug-resistant parasites. Exploration of novel antimalarial drug targets identified a trifunctional enzyme, malate quinone oxidoreductase (MQO), located in the mitochondrial inner membrane of P. falciparum (PfMQO). PfMQO is involved in the pathways of mitochondrial electron transport chain, tricarboxylic acid cycle, and fumarate cycle. Recent studies have shown that MQO is essential for P. falciparum survival in asexual stage and for the development of experiment cerebral malaria in the murine parasite P. berghei, providing genetic validation of MQO as a drug target. However, chemical validation of MQO, as a target, remains unexplored. In this study, we used active recombinant protein rPfMQO overexpressed in bacterial membrane fractions to screen a total of 400 compounds from the Pathogen Box, released by Medicines for Malaria Venture. The screening identified seven hit compounds targeting rPfMQO with an IC50 of under 5 µM. We tested the activity of hit compounds against the growth of 3D7 wildtype strain of P. falciparum, among which four compounds showed an IC50 from low to sub-micromolar concentrations, suggesting that PfMQO is indeed a potential antimalarial drug target.


Subject(s)
Enzyme Inhibitors/pharmacology , Malaria, Cerebral/drug therapy , Malaria, Falciparum/drug therapy , Oxidoreductases/antagonists & inhibitors , Animals , Antimalarials/metabolism , Antimalarials/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Malaria, Cerebral/enzymology , Malaria, Cerebral/parasitology , Malaria, Falciparum/enzymology , Malaria, Falciparum/parasitology , Malates/metabolism , Mice , Mitochondria/enzymology , Oxidoreductases/genetics , Plasmodium berghei/drug effects , Plasmodium berghei/pathogenicity , Plasmodium falciparum/enzymology , Plasmodium falciparum/pathogenicity , Quinones/metabolism
10.
Neurosci Lett ; 662: 290-294, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29107705

ABSTRACT

Cerebral malaria (CM) is a severe form of malaria caused by Plasmodium falciparum and P.vivax. CM affects the brain leading to coma and is the leading cause of death in malaria patients. The enzyme, adenosine 5'-monophosphate-activated protein kinase (AMPK), is an important metabolic sensor that helps in maintaining energy homeostasis during normal physiological as well as pathological conditions. In the present study, we studied the status of AMPK in the mouse model of CM. The C57BL/6 mice infected by rodent-specific P.berghei ANKA were used for the study. We found a statistically significant reduction in the gene expressions of Prkaa1 (α1 subunit) and Prkaa2 (α2 subunit) in the brains of CM mice compared to uninfected control. Also, there was a statistically significant reduction in the ratio of phospho-AMPK/AMPK protein levels in CM compared to uninfected control. There was no statistically significant decrease in phospho-ACC/ACC ratio in the brain compared to control. As AMPK is downregulated in CM, there is a possible involvement in neuronal cell death during CM pathogenesis, and therefore we feel that novel AMPK activating drugs might be helpful as an adjunctive therapy for conferring neuroprotection.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Brain/enzymology , Malaria, Cerebral/enzymology , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Plasmodium berghei
11.
Mol Med Rep ; 16(2): 2045-2050, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28656241

ABSTRACT

Hydrogen sulfide (H2S) has anti­inflammatory and neuroprotective properties, particularly during pathological processes. Experimental cerebral malaria (ECM), which is caused by vascular leakage into the brain, is characterized by inflammation, neurological deficits and cerebral hemorrhage. The present study investigated the correlation between ECM genesis and the levels of H2S. The results indicated that the levels of H2S derived from the brain decreased over time following ECM infection, and that the low H2S bioavailability was partially caused by decreased expression of the H2S generating enzyme, cystathionine­ß­synthase. Administration of NaHS (an exogenous donor of H2S) provided protection against ECM. NaHS inhibited the destruction of the blood brain barrier and the secretion of proinflammatory biomarkers, including interluekin­18, matrix metalloproteinase­9 and serum cluster of differentiation 40 into the brain during ECM. In conclusion, these results suggested that low levels of H2S in brain contributed to the progression of ECM, and that H2S donor administration may represent a potential protective therapy against ECM.


Subject(s)
Hydrogen Sulfide/therapeutic use , Malaria, Cerebral/drug therapy , Neuroprotective Agents/therapeutic use , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Cystathionine beta-Synthase/metabolism , Disease Models, Animal , Female , Hydrogen Sulfide/pharmacology , Inflammation/pathology , Malaria, Cerebral/blood , Malaria, Cerebral/enzymology , Malaria, Cerebral/pathology , Male , Matrix Metalloproteinase 9/blood , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Reproducibility of Results , Survival Analysis
12.
Neurochem Int ; 48(2): 108-13, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16236382

ABSTRACT

Neuronal calpains appear to be activated uncontrollably by sustained elevation of cytosolic calcium levels under pathological conditions as well as neurodegenerative diseases. In the present study, we have characterized calpain activation in cytosolic extract of mice cerebral cortex and cerebellum using an experimental model of fatal murine cerebral malaria (FMCM). Pathology of FMCM resulted in the increase in activity of calpains in both cerebral cortex and cerebellum. Western blot analysis revealed an increase in the levels of mu-calpain (calpain-1) in the cytosolic fraction of infected cerebral cortex and cerebellum although a decrease in the level of m-calpain was observed in the cytosolic fraction of infected cerebellum and cerebral cortex. Calpain activation was further confirmed by monitoring the formation of calpain-specific spectrin breakdown products (SBDP). Protease-specific SBDP revealed the formation of calpain-generated 150kDa product in the infected cerebral cortex and cerebellum. The specific signature fragment of calpain activation and spectrin breakdown after Plasmodium berghei ANKA infection provide a strong evidence of the role of calpains during the cell death in cerebral cortex and cerebellum. Given the role of calpains in neurodegeneration and cell death, our results strongly suggest that calpains are important mediators of cell injury and neurological sequelae associated with FMCM.


Subject(s)
Brain/metabolism , Calcium-Binding Proteins/metabolism , Calpain/metabolism , Malaria, Cerebral/metabolism , Spectrin/metabolism , Animals , Blotting, Western , Brain/enzymology , Enzyme Activation , Female , Hydrolysis , Immunohistochemistry , Malaria, Cerebral/enzymology , Malaria, Cerebral/pathology , Male , Mice
13.
Jpn J Infect Dis ; 58(5): 268-71, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16249618

ABSTRACT

Cerebral malaria (CM) is a serious complication of Plasmodium falciparum malaria, and its pathogenesis leading to coma remains unknown. Heme oxygenase-1 (HO-1) catalyzes heme breakdown, eventually generating bilirubin, iron and carbon monoxide. The HO-1 gene promoter contains a polymorphic (GT)n repeat which may influence the expression level of HO-1. To explore the correlation between this (GT)n polymorphism and susceptibility to CM, we analyzed the frequencies of the (GT)n alleles in 120 Myanmarese patients with uncomplicated malaria (UM) and 30 patients with CM. The frequency of homozygotes for the short (GT)n alleles (<28 repeats) in CM patients was significantly higher than those in UM patients (P < 0.008, OR = 3.14). Thus, short (GT)n alleles represent a genetic risk factor for CM.


Subject(s)
Heme Oxygenase-1/genetics , Malaria, Cerebral/enzymology , Malaria, Cerebral/genetics , Adult , Alleles , Dinucleotide Repeats , Homozygote , Humans , Malaria, Cerebral/etiology , Malaria, Falciparum/enzymology , Malaria, Falciparum/genetics , Myanmar , Polymorphism, Genetic , Promoter Regions, Genetic
14.
J Commun Dis ; 37(1): 44-50, 2005 Mar.
Article in English | MEDLINE | ID: mdl-16637400

ABSTRACT

Human cerebral malaria is caused by a protozoan parasitic with no cure till date. The isolation of brain capillaries i.e. microvessels has permitted the in vitro study related to cerebral function. Microvessels were isolated from normal and P. yoelii infected mice brain cortex and subjected to biochemical characterization by the following enzyme markers viz alkaline phosphatase, gamma-glutamyI transpeptidase and monoamine oxidase and electron microscopically. Limited studies have been carried out in relation to drug metabolizing enzymes in cerebral microvessels of rodents. The present studies have been carried out in relation to status of drug metabolizing enzymes during P. yoelii infection in cerebral microvessels of mice. The data obtained depicted a clear cut impairment of cytochrome P450 (a terminal monooxygenase) and related indices viz b5, benzopyrene hydroxylase, aminopyrene-n-demethylase, aniline hydroxylase except NADH cytochrome e reductase which increased during P. yoelii infection in mice as compared to normal. Further the oral drug administration (arteether) treatment brought back the altered MFO system normal a week alter cessation of drug treatment.


Subject(s)
Antimalarials/therapeutic use , Artemisinins/therapeutic use , Brain/enzymology , Malaria, Cerebral/drug therapy , Muscle, Smooth, Vascular/enzymology , Animals , Disease Models, Animal , Malaria, Cerebral/enzymology , Mice , Muscle, Smooth, Vascular/drug effects , Plasmodium yoelii
15.
PLoS One ; 10(3): e0119633, 2015.
Article in English | MEDLINE | ID: mdl-25775137

ABSTRACT

Experimental cerebral malaria (ECM) is characterized by a strong immune response, with leukocyte recruitment, blood-brain barrier breakdown and hemorrhage in the central nervous system. Phosphatidylinositol 3-kinase γ (PI3Kγ) is central in signaling diverse cellular functions. Using PI3Kγ-deficient mice (PI3Kγ-/-) and a specific PI3Kγ inhibitor, we investigated the relevance of PI3Kγ for the outcome and the neuroinflammatory process triggered by Plasmodium berghei ANKA (PbA) infection. Infected PI3Kγ-/- mice had greater survival despite similar parasitemia levels in comparison with infected wild type mice. Histopathological analysis demonstrated reduced hemorrhage, leukocyte accumulation and vascular obstruction in the brain of infected PI3Kγ-/- mice. PI3Kγ deficiency also presented lower microglial activation (Iba-1+ reactive microglia) and T cell cytotoxicity (Granzyme B expression) in the brain. Additionally, on day 6 post-infection, CD3+CD8+ T cells were significantly reduced in the brain of infected PI3Kγ-/- mice when compared to infected wild type mice. Furthermore, expression of CD44 in CD8+ T cell population in the brain tissue and levels of phospho-IkB-α in the whole brain were also markedly lower in infected PI3Kγ-/- mice when compared with infected wild type mice. Finally, AS605240, a specific PI3Kγ inhibitor, significantly delayed lethality in infected wild type mice. In brief, our results indicate a pivotal role for PI3Kγ in the pathogenesis of ECM.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/genetics , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Malaria, Cerebral/immunology , Malaria, Cerebral/pathology , Plasmodium berghei/immunology , Animals , Brain/immunology , Disease Models, Animal , Extracellular Matrix/immunology , Extracellular Matrix/parasitology , Female , Humans , Lung/enzymology , Lung/parasitology , Malaria, Cerebral/enzymology , Malaria, Cerebral/parasitology , Mice , Phosphoinositide-3 Kinase Inhibitors , Quinoxalines/pharmacology , Survival Analysis , Thiazolidinediones/pharmacology
16.
J Neuroimmunol ; 106(1-2): 198-205, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10814798

ABSTRACT

Intravascular sequestration and altered cytokine expression patterns are key determinators of CNS lesion formation in patients with cerebral malaria (CM). Among others, altered prostaglandin concentrations were revealed by clinical trials in peripheral blood of CM patients. Prostaglandin synthesis is controlled by cyclooxygenases (COX, prostaglandin endoperoxide synthase, PGG/H synthase) and COX expression has been attributed a key role in immunomodulation, hemostasis and inflammation in a wide variety of pathologically altered brain tissues. We have now analyzed expression of COX-1 and COX-2 in brains of patients with CM by immunohistochemistry. Double labeling experiments were used to verify the cellular identity of COX-1 and COX-2 expressing cells. Compared to healthy controls, significant (P=0.0006) accumulation of COX-1 expressing macrophages/microglial cells was detected in Dürck's granulomas. Accumulations of COX-2 expressing endothelial cells (P=0.0006) and COX-2 expressing astrocytes (P=0.0012) were detected in CM brain parenchyma. The restricted expression and accumulation of COX-1 and COX-2 in CM brains adds convincing evidence for the participation of cyclooxygenases in the formation of fever, inflammation and granuloma in these patients.


Subject(s)
Brain/enzymology , Isoenzymes/metabolism , Malaria, Cerebral/enzymology , Neurons/enzymology , Prostaglandin-Endoperoxide Synthases/metabolism , Brain/pathology , Cyclooxygenase 1 , Cyclooxygenase 2 , Humans , Immunohistochemistry , Malaria, Cerebral/pathology , Membrane Proteins , Reference Values
17.
J Neuroimmunol ; 142(1-2): 101-11, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14512169

ABSTRACT

In cerebral malaria (CM), microvascular activation accompanies blood-brain barrier dysfunction which in turn represents the pathophysiological basis of neurological impairments in affected patients. To dissect the molecular basis of this process, we analyzed localization of proangiogenic vascular endothelial growth factor (VEGF), its receptor vascular endothelial growth factor receptor-1 (VEGFR-1, Flt-1), of downstream VEGF effectors matrix-metalloproteinase-1 (MMP-1) and connective tissue growth factor (CTGF), and of VEGF-interacting antiangiogenic thrombospondin-1 and -independent angiostatin in brains of patients who died with CM and controls by immunohistochemistry and Western blotting experiments. Most prominently, we detected more VEGF(+) astrocytes in CM patients and deposition of Flt-1 in Dürck's granulomas. MMP-1 and thrombospondin-1 accumulated in macrophages/microglial cells in Dürck's granulomas. In one CM patient, massive amounts of CTGF were detected as perivascular paracellular deposits. Angiostatin was observed in the serum of 2/7 control but in no CM patients. These data demonstrate the activation of the proangiogenic VEGF signaling cascade in patients with CM, probably reflecting compensatory mechanisms of general and focal brain hypoxia observed in these patients.


Subject(s)
Angiogenic Proteins/analysis , Brain Chemistry , Malaria, Cerebral/metabolism , Angiostatins/analysis , Blotting, Western , Connective Tissue Growth Factor , Fluorescent Antibody Technique , Humans , Immediate-Early Proteins/analysis , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/analysis , Malaria, Cerebral/enzymology , Malaria, Cerebral/mortality , Malaria, Cerebral/pathology , Matrix Metalloproteinase 1/analysis , Thrombospondin 1/analysis , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor Receptor-1/analysis
18.
Int J Parasitol ; 34(12): 1309-19, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15542091

ABSTRACT

Products of the kynurenine pathway of tryptophan metabolism have been implicated in the pathogenesis of murine and human cerebral malaria. Indoleamine 2,3-dioxygenase is the first and rate-limiting enzyme in this pathway and we have developed an immunohistochemical method for its detection in tissues from normal and malaria-infected mice. Mice were infected with Plasmodium berghei ANKA, a murine model of cerebral malaria, or P. berghei K173, a non-cerebral malaria model. Vascular endothelial cells were the primary sites of indoleamine 2,3-dioxygenase expression in both types of malaria infection and this response was systemic, with positive staining of vascular endothelium in all tissues examined. No indoleamine 2,3-dioxygenase expression was detected in uninfected or interferon-gamma-/- mice. Corroborative data were obtained using quantitative reverse transcription PCR for indoleamine 2,3-dioxygenase mRNA. These results suggest that interferon-gamma-dependent indoleamine 2,3-dioxygenase expression is part of a normal systemic host response to the parasite, perhaps performing some tissue protective functions that may become deranged under some circumstances and contribute to the pathogenesis of cerebral malaria. On the other hand, constitutive indoleamine 2,3-dioxygenase expression in the epididymis and the placenta was detected in both C57Bl/6 wild-type and interferon-gamma-/- mice, suggesting a distinct regulatory mechanism for its induction in these normal physiological situations. Although increased indoleamine 2,3-dioxygenase production during murine malaria infection may not by itself cause cerebral pathology, metabolites of the kynurenine pathway may combine with other features of cerebral malaria, such as breakdown of the blood-brain barrier, to influence CNS function and contribute to the symptoms and pathology observed.


Subject(s)
Endothelium, Vascular/enzymology , Malaria, Cerebral/enzymology , Plasmodium berghei , Tryptophan Oxygenase/analysis , Animals , Enzyme Activation , Female , Immunohistochemistry/methods , Interferon-gamma/genetics , Interferon-gamma/immunology , Malaria, Cerebral/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Models, Animal , Parasitemia , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Tryptophan Oxygenase/genetics
19.
Am J Trop Med Hyg ; 70(6): 645-50, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15211007

ABSTRACT

Lipid mediators play important roles in the pathogenesis of malaria. Phospholipase A2s are enzymes involved in the production of these mediators, and they function in inflammation. Among them, cytosolic phospholipase A2 (cPLA2) is a key enzyme in the metabolism of arachidonic acid, the first intermediate in the production of lipid mediators. Plasmodium berghei ANKA causes cerebral malaria in CL57B/6 mice, and we recently produced cPLA2-deficient mice with this background. With the expectation of reduced pathogenicity, we performed experimental infection in these mice. Unexpectedly, the infected mice developed cerebral malaria and died at the same time as the control mice, while the parasitemia progressed similarly in both groups. These observations suggest that secretory PLA2s rather than cPLA2 may be involved in the aggravation, although possible compensation by the induction of other enzymes has not been excluded. The present findings are expected to help clarify the involvement of various phospholipase A2s in malaria.


Subject(s)
Cytosol/enzymology , Malaria, Cerebral/mortality , Phospholipases A/genetics , Plasmodium berghei , Animals , Female , Group II Phospholipases A2 , Malaria, Cerebral/enzymology , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Parasitemia/enzymology , Parasitemia/mortality , Phospholipases A/metabolism , Phospholipases A2
20.
Am J Trop Med Hyg ; 49(4): 455-9, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8214274

ABSTRACT

Some clinical manifestations of severe malaria resemble those of sepsis and there may be mediators of the host response that are common to both sepsis and malaria. Phospholipase A2 (PLA2), a proinflammatory enzyme whose expression is induced by tumor necrosis factor (TNF), has been implicated in the pathogenesis of complications of the sepsis syndrome. We examined levels of circulating PLA2 in Plasmodium falciparum malaria and studied the association of PLA2 with disease severity. Plasma PLA2 and TNF were measured in 75 Malawian children with P. falciparum malaria. The mean (SD) plasma PLA2 activity in children with acute malaria was 53,804 (37,256) units/ml as compared with 424 (349) units/ml in 34 healthy controls (P < 0.00001). The mean PLA2 activity in 45 convalescent patients was 2,546 (7,372) units/ml (P < 0.00001). In 48 patients with pretreatment PLA2 activity less than 60,000 units/ml, mortality was 8.3%, while in 27 patients with pretreatment PLA2 levels greater than 60,000 units/ml, mortality was 33.3% (P = 0.008). There were significant correlations between PLA2 and TNF (r = 0.471, P < 0.01), density of parasitemia (r = 0.443, P < 0.0001) and a decrease in hematocrit (r = 0.352, P < 0.005). These data show that P. falciparum malaria is associated with a markedly increased circulating PLA2, especially in patients with severe disease, as manifested by high parasite burden, anemia, coma, and death.


Subject(s)
Malaria, Cerebral/enzymology , Malaria, Falciparum/enzymology , Phospholipases A/blood , Acute Disease , Anemia/enzymology , Anemia/etiology , Animals , Child , Child, Preschool , Coma/enzymology , Coma/etiology , Female , Follow-Up Studies , Hematocrit , Humans , Infant , Malaria, Cerebral/blood , Malaria, Cerebral/complications , Malaria, Cerebral/mortality , Malaria, Falciparum/blood , Malaria, Falciparum/complications , Malaria, Falciparum/mortality , Malawi , Male , Nervous System Diseases/enzymology , Nervous System Diseases/etiology , Phospholipases A/cerebrospinal fluid , Phospholipases A2 , Plasmodium falciparum/enzymology , Tumor Necrosis Factor-alpha/analysis
SELECTION OF CITATIONS
SEARCH DETAIL