Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 239
Filter
Add more filters

Publication year range
1.
J Infect Dis ; 230(2): e474-e485, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-38271704

ABSTRACT

BACKGROUND: Transcriptomics has been used to evaluate immune responses during malaria in diverse cohorts worldwide. However, the high heterogeneity of cohorts and poor generalization of transcriptional signatures reported in each study limit their potential clinical applications. METHODS: We compiled 28 public data sets containing 1556 whole-blood or peripheral blood mononuclear cell transcriptome samples. We estimated effect sizes with Hedge's g value and the DerSimonian-Laird random-effects model for meta-analyses of uncomplicated malaria. Random forest models identified gene signatures that discriminate malaria from bacterial infections or malaria severity. Parasitological, hematological, immunological, and metabolomics data were used for validation. RESULTS: We identified 3 gene signatures: the uncomplicated Malaria Meta-Signature, which discriminates Plasmodium falciparum malaria from uninfected controls; the Malaria or Bacteria Signature, which distinguishes malaria from sepsis and enteric fever; and the cerebral Malaria Meta-Signature, which characterizes individuals with cerebral malaria. These signatures correlate with clinical hallmark features of malaria. Blood transcription modules indicate immune regulation by glucocorticoids, whereas cell development and adhesion are associated with cerebral malaria. CONCLUSIONS: Transcriptional meta-signatures reflecting immune cell responses provide potential biomarkers for translational innovation and suggest critical roles for metabolic regulators of inflammation during malaria.


Subject(s)
Biomarkers , Malaria, Falciparum , Plasmodium falciparum , Malaria, Falciparum/diagnosis , Malaria, Falciparum/parasitology , Humans , Biomarkers/blood , Plasmodium falciparum/genetics , Transcriptome , Gene Expression Profiling , Malaria, Cerebral/diagnosis , Malaria, Cerebral/genetics , Malaria, Cerebral/blood , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/immunology
2.
Malar J ; 23(1): 253, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39180112

ABSTRACT

BACKGROUND: Disordered amino acid metabolism is observed in cerebral malaria (CM). This study sought to determine whether abnormal amino acid concentrations were associated with level of consciousness in children recovering from coma. Twenty-one amino acids and coma scores were quantified longitudinally and the data were analysed for associations. METHODS: In a prospective observational study, 42 children with CM were enrolled. Amino acid levels were measured at entry and at frequent intervals thereafter and consciousness was assessed by Blantyre Coma Scores (BCS). Thirty-six healthy children served as controls for in-country normal amino acid ranges. Logistic regression was employed using a generalized linear mixed-effects model to assess associations between out-of-range amino acid levels and BCS. RESULTS: At entry 16/21 amino acid levels were out-of-range. Longitudinal analysis revealed 10/21 out-of-range amino acids were significantly associated with BCS. Elevated phenylalanine levels showed the highest association with low BCS. This finding held when out-of-normal-range data were analysed at each sampling time. CONCLUSION: Longitudinal data is provided for associations between abnormal amino acid levels and recovery from CM. Of 10 amino acids significantly associated with BCS, elevated phenylalanine may be a surrogate for impaired clearance of ether lipid mediators of inflammation and may contribute to CM pathogenesis.


Subject(s)
Amino Acids , Coma , Malaria, Cerebral , Humans , Coma/blood , Amino Acids/blood , Malaria, Cerebral/blood , Malaria, Cerebral/complications , Female , Male , Prospective Studies , Child, Preschool , Longitudinal Studies , Infant , Child
3.
Int J Mol Sci ; 25(17)2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39273566

ABSTRACT

Cerebral malaria in young African children is associated with high mortality, and persisting neurological deficits often remain in survivors. Sequestered Plasmodium-infected red blood cells lead to cerebrovascular inflammation and subsequent neuroinflammation. Brain inflammation can play a role in the pathogenesis of neurologic sequelae. Therefore, we assessed a select set of proinflammatory analytes (IP10, IL23, MIP3α, GRO, MCP-1, and osteopontin in both the plasma and cerebrospinal fluid(CSF) of Zambian children with cerebral malaria and compared this with children with neurological symptoms that were negative for Plasmodium falciparum (non-cerebral malaria). Several similarities in plasma and CSF levels were found, as were some striking differences. We confirmed that IP10 levels were higher in the plasma of cerebral malaria patients, but this was not found in CSF. Levels of osteopontin were elevated in both the plasma and CSF of CM patients compared to the non-CM patients. These results show again a highly inflammatory environment in both groups but a different profile for CM when compared to non-cerebral malaria. Osteopontin may play an important role in neurological inflammation in CM and the resulting sequelae. Therefore, osteopontin could be a valid target for further biomarker research and potentially for therapeutic interventions in neuroinflammatory infections.


Subject(s)
Biomarkers , Malaria, Cerebral , Osteopontin , Humans , Malaria, Cerebral/cerebrospinal fluid , Malaria, Cerebral/blood , Malaria, Cerebral/parasitology , Osteopontin/cerebrospinal fluid , Osteopontin/blood , Male , Female , Child, Preschool , Biomarkers/cerebrospinal fluid , Biomarkers/blood , Inflammation/cerebrospinal fluid , Inflammation/blood , Child , Plasmodium falciparum/pathogenicity , Infant , Malaria, Falciparum/cerebrospinal fluid , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology
4.
Malar J ; 19(1): 167, 2020 Apr 26.
Article in English | MEDLINE | ID: mdl-32336276

ABSTRACT

BACKGROUND: Cerebral malaria (CM), is a life-threatening childhood malaria syndrome with high mortality. CM is associated with impaired consciousness and neurological damage. It is not fully understood, as yet, why some children develop CM. Presented here is an observation from longitudinal studies on CM in a paediatric cohort of children from a large, densely-populated and malaria holoendemic, sub-Saharan, West African metropolis. METHODS: Plasma samples were collected from a cohort of children with CM, severe malarial anaemia (SMA), uncomplicated malaria (UM), non-malaria positive healthy community controls (CC), and coma and anemic patients without malaria, as disease controls (DC). Proteomic two-dimensional difference gel electrophoresis (2D-DIGE) and mass spectrometry were used in a discovery cohort to identify plasma proteins that might be discriminatory among these clinical groups. The circulatory levels of identified proteins of interest were quantified by ELISA in a prospective validation cohort. RESULTS: The proteome analysis revealed differential abundance of circulatory complement-lysis inhibitor (CLI), also known as Clusterin (CLU). CLI circulatory level was low at hospital admission in all children presenting with CM and recovered to normal level during convalescence (p < 0.0001). At acute onset, circulatory level of CLI in the CM group significantly discriminates CM from the UM, SMA, DC and CC groups. CONCLUSIONS: The CLI circulatory level is low in all patients in the CM group at admission, but recovers through convalescence. The level of CLI at acute onset may be a specific discriminatory marker of CM. This work suggests that CLI may play a role in the pathophysiology of CM and may be useful in the diagnosis and follow-up of children presenting with CM.


Subject(s)
Clusterin/blood , Convalescence , Malaria, Cerebral/parasitology , Malaria, Falciparum/parasitology , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Malaria, Cerebral/blood , Malaria, Falciparum/blood , Male , Prospective Studies
5.
BMC Infect Dis ; 20(1): 442, 2020 Jun 23.
Article in English | MEDLINE | ID: mdl-32576141

ABSTRACT

BACKGROUND: Cerebral malaria is the most severe form of infection with Plasmodium falciparum characterized by a highly inflammatory response. This systematic review aimed to investigate the association between TNF-α levels and cerebral malaria. METHODS: This review followed the Preferred Reporting of Systematic Review and Meta-analyses (PRISMA) guidelines. The search was performed at PubMed, LILACS, Scopus, Web of Science, The Cochrane Library, OpenGrey and Google Scholar. We have included studies of P. falciparum-infected humans with or without cerebral malaria and TNF-α dosage level. All studies were evaluated using a risk of bias tool and the GRADE approach. RESULTS: Our results have identified 2338 studies, and 8 articles were eligible according to this systematic review inclusion criteria. Among the eight articles, five have evaluated TNF- α plasma dosage, while two have evaluated at the blood and one at the brain (post-Morten). Among them, only five studies showed higher TNF-α levels in the cerebral malaria group compared to the severe malaria group. Methodological problems were identified regarding sample size, randomization and blindness, but no risk of bias was detected. CONCLUSION: Although the results suggested that that TNF-α level is associated with cerebral malaria, the evidence is inconsistent and imprecise. More observational studies evaluating the average TNF-alpha are needed.


Subject(s)
Malaria, Cerebral/epidemiology , Malaria, Falciparum/epidemiology , Tumor Necrosis Factor-alpha/blood , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Humans , Malaria, Cerebral/blood , Malaria, Falciparum/blood , Male , Middle Aged , Plasmodium falciparum , Young Adult
6.
Blood ; 127(19): 2281-8, 2016 05 12.
Article in English | MEDLINE | ID: mdl-26851291

ABSTRACT

Severe Plasmodium falciparum malaria remains a leading cause of mortality, particularly in sub-Saharan Africa where it accounts for up to 1 million deaths per annum. In spite of the significant mortality and morbidity associated with cerebral malaria (CM), the molecular mechanisms involved in the pathophysiology of severe malaria remain surprisingly poorly understood. Previous studies have demonstrated that sequestration of P falciparum-infected erythrocytes within the microvasculature of the brain plays a key role in the development of CM. In addition, there is convincing evidence that both endothelial cell activation and platelets play critical roles in the modulating the pathogenesis of severe P falciparum malaria. In this review, we provide an overview of recent studies that have identified novel roles through which hemostatic dysfunction may directly influence malaria pathogenesis. In particular, we focus on emerging data suggesting that von Willebrand factor, coagulation cascade activation, and dysfunction of the protein C pathway may be of specific importance in this context. These collective insights underscore a growing appreciation of the important, but poorly understood, role of hemostatic dysfunction in malaria progression and, importantly, illuminate potential approaches for novel therapeutic strategies. Given that the mortality rate associated with CM remains on the order of 20% despite the availability of effective antimalarial therapy, development of adjunctive therapies that can attenuate CM progression clearly represents a major unmet need. These emerging data are thus not only of basic scientific interest, but also of direct clinical significance.


Subject(s)
Blood Coagulation Disorders , Blood Coagulation , Malaria, Falciparum , Plasmodium falciparum , Blood Coagulation Disorders/blood , Blood Coagulation Disorders/etiology , Blood Coagulation Disorders/pathology , Blood Coagulation Disorders/therapy , Blood Platelets/metabolism , Blood Platelets/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Erythrocytes/metabolism , Erythrocytes/pathology , Humans , Malaria, Cerebral/blood , Malaria, Cerebral/complications , Malaria, Cerebral/pathology , Malaria, Cerebral/therapy , Malaria, Falciparum/blood , Malaria, Falciparum/complications , Malaria, Falciparum/pathology , Malaria, Falciparum/therapy , Protein C/metabolism , von Willebrand Factor/metabolism
7.
Microb Pathog ; 107: 69-74, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28336326

ABSTRACT

The development and spread of multidrug-resistant strains of malarial parasites have led to an overwhelming increase in the resistance to current antimalarial drugs. The urgent need for alternative antimalarial drugs has directed some of the current studies toward folkloric medicine approaches. Interestingly, the Zizyphus spina Cristi leaf extract (ZLE) has been found to exhibit antiplasmodial activity. This study evaluated the protective effect of ZLE against Plasmodium berghei-induced cerebral tissue injuries in mice. Male C57Bl/6 mice received an injection of P. berghei-infected red blood cells. Mice were divided into three groups (control, infected, and ZLE-treated), and were subjected to histological, biochemical, and molecular analyses. Murine malaria infections induced significant weight loss; however, upon ZLE treatment, the weight of mice was markedly restored. Additionally, infected mice showed brain histopathological changes and induction of oxidative damage. Significantly, ZLE treatment restored the levels of oxidative markers and antioxidant enzyme to the normal ranges. The mRNA expression of several genes in the brain of mice including Cacnb4, Adam23, Glrb, Vdac3, and Cabp1 was significantly upregulated during P. berghei infection. In contrast, ZLE markedly reduced the mRNA expression of these genes. To conclude, the results indicate that ZLE could play an important role in reducing the destructive effect of P. berghei-induced cerebral malaria owing to its antiplasmodial and antioxidant activities.


Subject(s)
Antimalarials/pharmacology , Gene Expression Regulation/drug effects , Malaria, Cerebral/drug therapy , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Ziziphus/chemistry , ADAM Proteins/genetics , Animals , Antioxidants , Brain/pathology , Brain/physiopathology , Calcium Channels/genetics , Calcium-Binding Proteins/genetics , Disease Models, Animal , Malaria/drug therapy , Malaria/parasitology , Malaria, Cerebral/blood , Malaria, Cerebral/pathology , Male , Mice , Mice, Inbred C57BL , Mitochondrial Membrane Transport Proteins/genetics , Nerve Tissue Proteins/genetics , Plant Leaves/chemistry , Plasmodium berghei/drug effects , Plasmodium berghei/pathogenicity , RNA, Messenger/drug effects , RNA, Messenger/genetics , Receptors, Glycine/genetics , Up-Regulation , Voltage-Dependent Anion Channels/genetics
8.
Transfusion ; 57(4): 985-988, 2017 04.
Article in English | MEDLINE | ID: mdl-28185296

ABSTRACT

BACKGROUND: Adjunctive automated whole blood or red blood cell exchange (RBCEx) can rapidly decrease malarial hyperparasitemia. Several case reports and series suggest improvement in clinical symptomatology; however, recent Centers of Disease Control and Prevention (CDC) recommendations concluded that RBCEx has no efficacy as an adjunctive therapy. We present a case of mental status changes secondary to cerebral malaria treated with automated RBCEx resulting in rapid and dramatic neurologic improvement. CASE REPORT: An 84-year-old Somali woman presented with a 3-day history of altered mental status, spiking fevers, chills, bilateral leg pain and weakness, and intermittent diarrhea. Her travel history included a recent trip to Kenya for 1 month without antimalarial chemoprophylaxis. During the hospital stay, her health declined, and she became obtunded. Physical examination revealed fever, tachypnea, hypertension, hypoxia, and no response to verbal or physical stimuli. Her hemoglobin decreased from 12.6 to 6.5 g/dL with 12% intraerythrocytic parasitemia by thin smear. Intraerythrocytic trophozoites and banana-shaped gametocytes were present consistent with Plasmodium falciparum. An emergent 1.5-volume RBC mass automated RBCEx and quinidine infusion decreased her parasitemia to 2%. The patient's mental status improved throughout the procedure, and after the 2½-hour procedure, the patient was alert, oriented, and speaking coherently. The patient continued to receive quinidine and artesunate 1 day later from CDC. CONCLUSION: Automated RBCEx transfusion reduced the parasite burden and restored neurologic functioning in a patient with cerebral malaria while awaiting definitive treatment with artesunate.


Subject(s)
Erythrocyte Transfusion , Malaria, Cerebral , Malaria, Falciparum , Parasitemia , Plasmodium falciparum , Quinidine/administration & dosage , Aged, 80 and over , Female , Humans , Malaria, Cerebral/blood , Malaria, Cerebral/parasitology , Malaria, Cerebral/therapy , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Malaria, Falciparum/therapy , Parasitemia/blood , Parasitemia/parasitology , Parasitemia/therapy
9.
J Infect Dis ; 213(1): 80-9, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26347576

ABSTRACT

Plasmodium falciparum-induced severe malaria remains a continuing problem in areas of endemicity, with elevated morbidity and mortality. Drugs targeting mechanisms involved in severe malaria pathology, including cytoadhesion of infected red blood cells (RBCs) to host receptors and production of proinflammatory cytokines, are still necessary. Human C1-inhibitor (C1INH) is a multifunctional protease inhibitor that regulates coagulation, vascular permeability, and inflammation, with beneficial effects in inflammatory disease models, including septic shock. We found that human C1INH, at therapeutically relevant doses, blocks severe malaria pathogenic processes by 2 distinct mechanisms. First, C1INH bound to glycan moieties within P. falciparum glycosylphosphatidylinositol (PfGPI) molecules on the parasite surface, inhibiting parasite RBC invasion and proinflammatory cytokine production by parasite-stimulated monocytes in vitro and reducing parasitemia in a rodent model of experimental cerebral malaria (ECM) in vivo. Second, C1INH bound to host CD36 and chondroitin sulfate A molecules, interfering with cytoadhesion of infected RBCs by competitive binding to these receptors in vitro and reducing sequestration in specific tissues and protecting against ECM in vivo. This study reveals that C1INH is a potential therapeutic antimalarial molecule able to interfere with severe-disease etiology at multiple levels through specific interactions with both parasite PfGPIs and host cell receptors.


Subject(s)
Cell Adhesion/drug effects , Complement C1 Inactivator Proteins/metabolism , Complement C1 Inactivator Proteins/pharmacology , Glycosylphosphatidylinositols/metabolism , Host-Parasite Interactions/drug effects , Malaria, Cerebral/metabolism , Malaria, Cerebral/parasitology , Protozoan Proteins/metabolism , Animals , Cell Line, Tumor , Complement C1 Inhibitor Protein , Disease Models, Animal , Erythrocytes/parasitology , Female , Humans , Malaria, Cerebral/blood , Mice , Mice, Inbred C57BL , Plasmodium berghei/metabolism , Plasmodium berghei/pathogenicity , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology
10.
PLoS Pathog ; 10(4): e1004038, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24743550

ABSTRACT

Systemic inflammation and sequestration of parasitized erythrocytes are central processes in the pathophysiology of severe Plasmodium falciparum childhood malaria. However, it is still not understood why some children are more at risks to develop malaria complications than others. To identify human proteins in plasma related to childhood malaria syndromes, multiplex antibody suspension bead arrays were employed. Out of the 1,015 proteins analyzed in plasma from more than 700 children, 41 differed between malaria infected children and community controls, whereas 13 discriminated uncomplicated malaria from severe malaria syndromes. Markers of oxidative stress were found related to severe malaria anemia while markers of endothelial activation, platelet adhesion and muscular damage were identified in relation to children with cerebral malaria. These findings suggest the presence of generalized vascular inflammation, vascular wall modulations, activation of endothelium and unbalanced glucose metabolism in severe malaria. The increased levels of specific muscle proteins in plasma implicate potential muscle damage and microvasculature lesions during the course of cerebral malaria.


Subject(s)
Malaria, Cerebral/blood , Oxidative Stress , Plasmodium falciparum , Proteomics/methods , Adolescent , Biomarkers/blood , Child , Child, Preschool , Female , Humans , Male , Syndrome
11.
PLoS Pathog ; 10(3): e1003839, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24651155

ABSTRACT

In patients with cerebral malaria (CM), higher levels of cell-specific microparticles (MP) correlate with the presence of neurological symptoms. MP are submicron plasma membrane-derived vesicles that express antigens of their cell of origin and phosphatidylserine (PS) on their surface, facilitating their role in coagulation, inflammation and cell adhesion. In this study, the in vivo production, fate and pathogenicity of cell-specific MP during Plasmodium berghei infection of mice were evaluated. Using annexin V, a PS ligand, and flow cytometry, analysis of platelet-free plasma from infected mice with cerebral involvement showed a peak of MP levels at the time of the neurological onset. Phenotypic analyses showed that MP from infected mice were predominantly of platelet, endothelial and erythrocytic origins. To determine the in vivo fate of MP, we adoptively transferred fluorescently labelled MP from mice with CM into healthy or infected recipient mice. MP were quickly cleared following intravenous injection, but microscopic examination revealed arrested MP lining the endothelium of brain vessels of infected, but not healthy, recipient mice. To determine the pathogenicity of MP, we transferred MP from activated endothelial cells into healthy recipient mice and this induced CM-like brain and lung pathology. This study supports a pathogenic role for MP in the aggravation of the neurological lesion and suggests a causal relationship between MP and the development of CM.


Subject(s)
Cell-Derived Microparticles/pathology , Malaria, Cerebral/blood , Plasmodium berghei/pathogenicity , Adoptive Transfer , Animals , Disease Models, Animal , Female , Flow Cytometry , Fluorescent Antibody Technique , Mice , Mice, Inbred CBA , Virulence
12.
Blood ; 123(2): 163-7, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24246501

ABSTRACT

Of all the outcomes of Plasmodium falciparum infection, the coma of cerebral malaria (CM) is particularly deadly. Malariologists have long wondered how some patients develop this organ-specific syndrome. Data from two recent publications support a novel mechanism of CM pathogenesis in which infected erythrocytes (IEs) express specific virulence proteins that mediate IE binding to the endothelial protein C receptor (EPCR). Malaria-associated depletion of EPCR, with subsequent impairment of the protein C system promotes a proinflammatory, procoagulant state in brain microvessels.


Subject(s)
Antigens, CD/metabolism , Erythrocytes/metabolism , Erythrocytes/parasitology , Malaria, Cerebral/metabolism , Malaria, Falciparum/metabolism , Plasmodium falciparum/metabolism , Receptors, Cell Surface/metabolism , Blood Coagulation , Cell Adhesion , Endothelial Protein C Receptor , Endothelium, Vascular/metabolism , Humans , Malaria, Cerebral/blood , Malaria, Cerebral/parasitology , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Microvessels , Organ Specificity , Plasmodium falciparum/genetics , Protein Binding
13.
J Infect Dis ; 211(9): 1484-8, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25425698

ABSTRACT

Loss of endothelial protein C receptor (EPCR) occurs at the sites of Plasmodium falciparum-infected erythrocyte sequestration in patients with or who died from cerebral malaria. In children presenting with different clinical syndromes of malaria, we assessed the relationships between endogenous plasma soluble EPCR (sEPCR) levels and clinical presentation or mortality. After adjustment for age, for treatment before admission, and for a known genetic factor, sEPCR level at admission was positively associated with cerebral malaria (P = .011) and with malaria-related mortality (P = .0003). Measuring sEPCR levels at admission could provide an early biological marker of the outcome of cerebral malaria.


Subject(s)
Antigens, CD/blood , Malaria, Cerebral/blood , Malaria, Cerebral/mortality , Receptors, Cell Surface/blood , Antigens, CD/metabolism , Antimalarials/therapeutic use , Benin/epidemiology , Child, Preschool , Endothelial Protein C Receptor , Genotype , Humans , Malaria, Cerebral/drug therapy , Malaria, Cerebral/epidemiology , Quinine/therapeutic use , Receptors, Cell Surface/metabolism
14.
PLoS Pathog ; 9(7): e1003491, 2013.
Article in English | MEDLINE | ID: mdl-23853600

ABSTRACT

Interferon Regulatory Factor 8 (IRF8) is required for development, maturation and expression of anti-microbial defenses of myeloid cells. BXH2 mice harbor a severely hypomorphic allele at Irf8 (Irf8(R294C)) that causes susceptibility to infection with intracellular pathogens including Mycobacterium tuberculosis. We report that BXH2 are completely resistant to the development of cerebral malaria (ECM) following Plasmodium berghei ANKA infection. Comparative transcriptional profiling of brain RNA as well as chromatin immunoprecipitation and high-throughput sequencing (ChIP-seq) was used to identify IRF8-regulated genes whose expression is associated with pathological acute neuroinflammation. Genes increased by infection were strongly enriched for IRF8 binding sites, suggesting that IRF8 acts as a transcriptional activator in inflammatory programs. These lists were enriched for myeloid-specific pathways, including interferon responses, antigen presentation and Th1 polarizing cytokines. We show that inactivation of several of these downstream target genes (including the Irf8 transcription partner Irf1) confers protection against ECM. ECM-resistance in Irf8 and Irf1 mutants is associated with impaired myeloid and lymphoid cells function, including production of IL12p40 and IFNγ. We note strong overlap between genes bound and regulated by IRF8 during ECM and genes regulated in the lungs of M. tuberculosis infected mice. This IRF8-dependent network contains several genes recently identified as risk factors in acute and chronic human inflammatory conditions. We report a common core of IRF8-bound genes forming a critical inflammatory host-response network.


Subject(s)
Brain/immunology , Gene Expression Regulation , Immunity, Innate , Interferon Regulatory Factors/metabolism , Malaria, Cerebral/immunology , Nerve Tissue Proteins/metabolism , Plasmodium berghei/immunology , Amino Acid Substitution , Animals , Binding Sites , Brain/metabolism , Brain/parasitology , Cells, Cultured , Cytokines/biosynthesis , Cytokines/blood , Gene Expression Profiling , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factors/chemistry , Interferon Regulatory Factors/genetics , Malaria, Cerebral/blood , Malaria, Cerebral/metabolism , Malaria, Cerebral/parasitology , Mice , Mice, Knockout , Mice, Mutant Strains , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurons/immunology , Neurons/metabolism , Neurons/parasitology , Specific Pathogen-Free Organisms , Spleen/immunology , Spleen/metabolism , Spleen/pathology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/parasitology
15.
Blood ; 121(15): 3016-22, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23380741

ABSTRACT

Cerebral malaria (CM) and severe malarial anemia (SMA) are the most serious life-threatening clinical syndromes of Plasmodium falciparum infection in childhood. Therefore, it is important to understand the pathology underlying the development of CM and SMA as opposed to uncomplicated malaria (UM). Increased levels of hepcidin have been associated with UM, but its level and role in severe malarial disease remains to be investigated. Plasma and clinical data were obtained as part of a prospective case-control study of severe childhood malaria at the main tertiary hospital of the city of Ibadan, Nigeria. Here, we report that hepcidin levels are lower in children with SMA or CM than in those with milder outcome (UM). While different profiles of pro- and anti-inflammatory cytokines were observed between the malaria syndromes, circulatory hepcidin levels remained associated with the levels of its regulatory cytokine interleukin-6 and of the anti-inflammatory cytokine inerleukin-10, irrespective of iron status, anemic status, and general acute-phase response. We propose a role for hepcidin in anti-inflammatory processes in childhood malaria.


Subject(s)
Antimicrobial Cationic Peptides/blood , Cytokines/blood , Inflammation Mediators/blood , Malaria, Cerebral/blood , Malaria, Falciparum/blood , Anemia/blood , Anemia/complications , Case-Control Studies , Child , Child, Preschool , Female , Ferritins/blood , Hematocrit , Hepcidins , Humans , Interleukin-10/blood , Interleukin-6/blood , Iron/blood , Linear Models , Malaria, Cerebral/complications , Malaria, Falciparum/complications , Male , Nigeria , Prospective Studies , Receptors, Transferrin/blood , Tertiary Care Centers , Transferrin/analysis
16.
Blood ; 122(5): 842-51, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23741007

ABSTRACT

Cerebral malaria (CM) is a major cause of mortality in African children and the mechanisms underlying its development, namely how malaria-infected erythrocytes (IEs) cause disease and why the brain is preferentially affected, remain unclear. Brain microhemorrhages in CM suggest a clotting disorder, but whether this phenomenon is important in pathogenesis is debated. We hypothesized that localized cerebral microvascular thrombosis in CM is caused by a decreased expression of the anticoagulant and protective receptors thrombomodulin (TM) and endothelial protein C receptor (EPCR) and that low constitutive expression of these regulatory molecules in the brain make it particularly vulnerable. Autopsies from Malawian children with CM showed cerebral fibrin clots and loss of EPCR, colocalized with sequestered IEs. Using a novel assay to examine endothelial phenotype ex vivo using subcutaneous microvessels, we demonstrated that loss of EPCR and TM at sites of IE cytoadherence is detectible in nonfatal CM. In contrast, although clotting factor activation was seen in the blood of CM patients, this was compensated and did not disseminate. Because of the pleiotropic nature of EPCR and TM, these data implicate disruption of the endothelial protective properties at vulnerable sites and particularly in the brain, linking coagulation and inflammation with IE sequestration.


Subject(s)
Antigens, CD/metabolism , Blood Coagulation/physiology , Brain/parasitology , Endothelium, Vascular/metabolism , Inflammation , Malaria, Cerebral/parasitology , Receptors, Cell Surface/metabolism , Antigens, CD/physiology , Black People , Blood Coagulation/immunology , Brain/blood supply , Brain/pathology , Case-Control Studies , Child , Child, Preschool , Down-Regulation , Endothelial Protein C Receptor , Erythrocytes/parasitology , Erythrocytes/pathology , Female , Humans , Infant , Inflammation/metabolism , Inflammation/parasitology , Malaria, Cerebral/blood , Malaria, Cerebral/immunology , Malaria, Cerebral/metabolism , Malawi , Male , Receptors, Cell Surface/physiology , Thrombomodulin/metabolism , Thrombomodulin/physiology
17.
J Immunol ; 190(9): 4685-91, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23536632

ABSTRACT

Platelets are most recognized as the cellular mediator of thrombosis, but they are increasingly appreciated for their immunomodulatory roles, including responses to Plasmodium infection. Platelet interactions with endothelial cells and leukocytes contribute significantly to the pathogenesis of experimental cerebral malaria (ECM). Recently, it has been suggested that platelets not only have an adverse role in cerebral malaria, but platelets may also be protective in animal models of uncomplicated malaria. We now demonstrate that these diverse and seemingly contradictory roles for platelets extend to cerebral malaria models and are dependent on the timing of platelet activation during infection. Our data show that platelets are activated very early in ECM and have a central role in initiation of the acute-phase response to blood-stage infection. Unlike platelet depletion or inhibition postinfection, preinfection platelet depletion or treatment with a platelet inhibitor is not protective. Additionally, we show that platelet-driven acute-phase responses have a major role in protecting mice from ECM by limiting parasite growth. Our data now suggest that platelets have a complex role in ECM pathogenesis: platelets help limit parasite growth early postinfection, but with continued platelet activation as the disease progresses, platelets contribute to ECM-associated inflammation.


Subject(s)
Acute-Phase Reaction/immunology , Blood Platelets/immunology , Malaria, Cerebral/blood , Malaria/blood , Platelet Activation/immunology , Animals , Disease Models, Animal , Malaria/immunology , Malaria, Cerebral/immunology , Mice , Mice, Inbred C57BL , Plasmodium berghei
18.
J Infect Dis ; 209(4): 600-9, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24041795

ABSTRACT

BACKGROUND: The diagnosis of cerebral malaria is problematic in malaria-endemic areas because encephalopathy in patients with parasitemia may have another cause. Abnormal retinal findings are thought to increase the specificity of the diagnosis, and the level of histidine-rich protein 2 (HRP2) may reflect the parasite biomass. METHODS: We examined the retina and measured plasma HRP2 levels in children with acute nontraumatic encephalopathy in Kenya. Logistic regression, with HRP2 level as an independent variable and World Health Organization-defined cerebral malaria and/or retinopathy as the outcome, was used to calculate malaria-attributable fractions (MAFs) and retinopathy-attributable fractions (RAFs). RESULTS: Of 270 children, 140 (52%) had peripheral parasitemia, 80 (30%) had malaria retinopathy, and 164 (61%) had an HRP2 level of >0 U/mL. During 2006-2011, the incidence of HRP2 positivity among admitted children declined by 49 cases per 100 000 per year (a 78% reduction). An HRP2 level of >0 U/mL had a MAF of 93% for cerebral malaria, with a MAF of 97% observed for HRP2 levels of ≥ 10 U/mL (the level of the best combined sensitivity and specificity). HRP2 levels of >0 U/mL had a RAF of 77% for features of retinopathy combined, with the highest RAFs for macular whitening (99%), peripheral whitening (98%), and hemorrhages (90%). CONCLUSION: HRP2 has a high attributable fraction for features of malarial retinopathy, supporting its use in the diagnosis of cerebral malaria. HRP2 thresholds improve the specificity of the definition.


Subject(s)
Antigens, Protozoan/blood , Malaria, Cerebral/blood , Malaria, Falciparum/blood , Protozoan Proteins/blood , Retinal Diseases/blood , Chi-Square Distribution , Child, Preschool , Diagnosis, Differential , Female , Humans , Incidence , Infant , Malaria, Cerebral/diagnosis , Malaria, Falciparum/diagnosis , Male , Prospective Studies , Retinal Diseases/epidemiology
19.
J Infect Dis ; 209(4): 610-5, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24048963

ABSTRACT

Endothelial dysregulation is central to the pathogenesis of acute Plasmodium falciparum infection. It has been assumed that this dysregulation resolves rapidly after treatment, but this return to normality has been neither demonstrated nor quantified. We therefore measured a panel of plasma endothelial markers acutely and in convalescence in Malawian children with uncomplicated or cerebral malaria. Evidence of persistent endothelial activation and inflammation, indicated by increased plasma levels of soluble intracellular adhesion molecule 1, angiopoetin 2, and C-reactive protein, were observed at 1 month follow-up visits. These vascular changes may represent a previously unrecognized contributor to ongoing malaria-associated morbidity and mortality.


Subject(s)
Endothelium/pathology , Malaria, Cerebral/pathology , Malaria, Falciparum/pathology , Analysis of Variance , Biomarkers/blood , Biomarkers/metabolism , C-Reactive Protein/metabolism , Case-Control Studies , Child, Preschool , Endothelium/metabolism , Female , Fever/blood , Fever/parasitology , Fever/pathology , Humans , Inflammation/blood , Inflammation/parasitology , Inflammation/pathology , Intercellular Adhesion Molecule-1/blood , Malaria, Cerebral/blood , Malaria, Falciparum/blood , Malawi , Male , Vesicular Transport Proteins/blood
20.
Infect Immun ; 82(3): 1287-95, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24379293

ABSTRACT

Nitric oxide (NO) is a proposed component of malaria pathogenesis, and the inducible nitric oxide synthase gene (NOS2) has been associated to malaria susceptibility. We analyzed the role of NOS2 polymorphisms on NO bioavailability and on susceptibility to infection, Plasmodium carrier status and clinical malaria. Two distinct West African sample collections were studied: a population-based collection of 1,168 apparently healthy individuals from the Príncipe Island and a hospital-based cohort of 269 Angolan children. We found that two NOS2 promoter single-nucleotide polymorphism (SNP) alleles associated to low NO plasma levels in noninfected individuals were also associated to reduced risk of pre-erythrocytic infection as measured anti-CSP antibody levels (6.25E-04 < P < 7.57E-04). In contrast, three SNP alleles within the NOS2 cistronic region conferring increased NO plasma levels in asymptomatic carriers were strongly associated to risk of parasite carriage (8.00E-05 < P < 7.90E-04). Notwithstanding, three SNP alleles in this region protected from cerebral malaria (7.90E-4 < P < 4.33E-02). Cohesively, the results revealed a dual regimen in the genetic control of NO bioavailability afforded by NOS2 depending on the infection status. NOS2 promoter variants operate in noninfected individuals to decrease both NO bioavailability and susceptibility to pre-erythrocytic infection. Conversely, NOS2 cistronic variants (namely, rs6505469) operate in infected individuals to increase NO bioavailability and confer increased susceptibility to unapparent infection but protect from cerebral malaria. These findings corroborate the hypothesis that NO anti-inflammatory properties impact on different steps of malaria pathogenesis, explicitly by favoring infection susceptibility and deterring severe malaria syndromes.


Subject(s)
Malaria, Cerebral/genetics , Malaria/genetics , Nitric Oxide Synthase Type II/genetics , Nitric Oxide/blood , Alleles , Biomarkers/blood , Humans , Malaria/blood , Malaria, Cerebral/blood , Plasmodium , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL