Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 177
1.
Eur J Med Res ; 28(1): 76, 2023 Feb 13.
Article En | MEDLINE | ID: mdl-36782344

BACKGROUND: Cerebral malaria (CM) is a manifestation of malaria caused by plasmodium infection. It has a high mortality rate and severe neurological sequelae, existing a significant research gap and requiring further study at the molecular level. METHODS: We downloaded the GSE117613 dataset from the Gene Expression Omnibus (GEO) database to determine the differentially expressed genes (DEGs) between the CM group and the control group. Weighted gene coexpression network analysis (WGCNA) was applied to select the module and hub genes most relevant to CM. The common genes of the key module and DEGs were selected to perform further analysis. The least absolute shrinkage and selection operator (LASSO) logistic regression and support vector machine recursive feature elimination (SVM-RFE) were applied to screen and verify the diagnostic markers of CM. Eventually, the hub genes were validated in the external dataset. Gene set enrichment analysis (GSEA) was applied to investigate the possible roles of the hub genes. RESULTS: The GO and KEGG results showed that DEGs were enriched in some neutrophil-mediated pathways and associated with some lumen structures. Combining LASSO and the SVM-RFE algorithms, LEF1 and IRAK3 were identified as potential hub genes in CM. Through the GSEA enrichment results, we found that LEF1 and IRAK3 participated in maintaining the integrity of the blood-brain barrier (BBB), which contributed to improving the prognosis of CM. CONCLUSIONS: This study may help illustrate the pathophysiology of CM at the molecular level. LEF1 and IRAK3 can be used as diagnostic biomarkers, providing new insight into the diagnosis and prognosis prediction in pediatric CM.


Malaria, Cerebral , Child , Humans , Malaria, Cerebral/diagnosis , Malaria, Cerebral/genetics , Africa , Algorithms , Machine Learning , Biomarkers
2.
PLoS One ; 18(1): e0280282, 2023.
Article En | MEDLINE | ID: mdl-36626386

Complement Receptor Type 1 (CR1) is a malaria-associated gene that encodes a transmembrane receptor of erythrocytes and is crucial for malaria parasite invasion. The expression of CR1 contributes to the rosetting of erythrocytes in the brain bloodstream, causing cerebral malaria, the most severe form of the disease. Here, we study the history of adaptation against malaria by analyzing selection signals in the CR1 gene. We used whole-genome sequencing datasets of 907 healthy individuals from malaria-endemic and non-endemic populations. We detected robust positive selection in populations from the hyperendemic regions of East India and Papua New Guinea. Importantly, we identified a new adaptive variant, rs12034598, which is associated with a slower rate of erythrocyte sedimentation and is linked with a variant associated with low levels of CR1 expression. The combination of the variants likely drives natural selection. In addition, we identified a variant rs3886100 under positive selection in West Africans, which is also related to a low level of CR1 expression in the brain. Our study shows the fine-resolution history of positive selection in the CR1 gene and suggests a population-specific history of CR1 adaptation to malaria. Notably, our novel approach using population genomic analyses allows the identification of protective variants that reduce the risk of malaria infection without the need for patient samples or malaria individual medical records. Our findings contribute to understanding of human adaptation against cerebral malaria.


Malaria, Cerebral , Receptors, Complement 3b , Humans , Erythrocytes , Malaria, Cerebral/genetics , Malaria, Cerebral/metabolism , Papua New Guinea , Receptors, Complement 3b/genetics , Selection, Genetic , Genetics, Population , India
3.
Malar J ; 21(1): 342, 2022 Nov 17.
Article En | MEDLINE | ID: mdl-36397106

BACKGROUND: Inducible expression of heme oxygenase-1 (encoded by the gene HMOX1) may determine protection from heme released during malaria infections. A variable length, short tandem GT(n) repeat (STR) in HMOX1 that may influence gene expression has been associated with outcomes of human malaria in some studies. In this study, an analysis of the association between variation at the STR in HMOX1 on severe malaria and severe malaria subtypes is presented in a large, prospectively collected dataset (MalariaGEN). METHODS: The HMOX1 STR was imputed using a recently developed reference haplotype panel designed for STRs. The STR was classified by total length and split into three alleles based on an observed trimodal distribution of repeat lengths. Logistic regression was used to assess the association between this repeat on cases of severe malaria and severe malaria subtypes (cerebral malaria and severe malarial anaemia). Individual analyses were performed for each MalariaGEN collection site and combined for meta-analysis. One site (Kenya), had detailed clinical metadata, allowing the assessment of the effect of the STR on clinical variables (e.g. parasite count, platelet count) and regression analyses were performed to investigate whether the STR interacted with any clinical variables. RESULTS: Data from 17,960 participants across 11 collection sites were analysed. In logistic regression, there was no strong evidence of association between STR length and severe malaria (Odds Ratio, OR: 0.96, 95% confidence intervals 0.91-1.02 per ten GT(n) repeats), although there did appear to be an association at some sites (e.g., Kenya, OR 0.90, 95% CI 0.82-0.99). There was no evidence of an interaction with any clinical variables. CONCLUSIONS: Meta-analysis suggested that increasing HMOX1 STR length is unlikely to be reliably associated with severe malaria. It cannot be ruled out that repeat length may alter risk in specific populations, although whether this is due to chance variation, or true variation due to underlying biology (e.g., gene vs environment interaction) remains unanswered.


Heme Oxygenase-1 , Malaria, Cerebral , Humans , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Genetic Predisposition to Disease , Polymorphism, Genetic , Alleles , Malaria, Cerebral/genetics
4.
Mol Omics ; 18(8): 716-730, 2022 09 26.
Article En | MEDLINE | ID: mdl-35960011

BACKGROUND: cerebral malaria (CM) is an important complication of malaria with a high mortality rate. Artesunate is recommended as the first-line artemisinin compound treatment for severe malaria. Due to the difficulty of obtaining brain tissue samples clinically, the use of animals to research host responses to CM parasite infections is necessary. Rodent malaria models allow for detailed time series studies of host responses in multiple organs. To date, studies on the transcriptome of severe malaria are only limited to the parasites in the peripheral blood of patients, and there is little data on the transcriptional changes in brain tissue in mice with CM treated with artesunate. METHOD AND RESULT: in this study, fresh tissue samples (three biological replicates per mouse) from the same area of the brain in each animal were collected from the uninfected, Plasmodium berghei ANKA-infected and artesunate-treated C57BL/6 mice, and then transcriptome research was performed by the RNA-seq technique. Differentially expressed genes (DEGs) included Il-21, Tnf, Il-6, Il-1ß, Il-10, Ifng, and Icam-1. Among which, Il-6, Il-10, Tnf-α and Il-1ß were further verified and validated via qRT-PCR and ELISA. This revealed that Il-1ß (p < 0.0001), Il-10 (p < 0.05) and Tnf-α (p < 0.05) were significantly up-regulated in the Pb ANKA-infected versus uninfected group, while Il-1ß (p < 0.0001) and Tnf-α (p < 0.05) were significantly down-regulated after artesunate treatment. All DEGs were closely related to the top 3 artesunate treatment pathways, including the JAK-STAT signaling pathway, apoptosis, and Toll-like receptor signaling pathway. CONCLUSION: the mechanism of improving the prognosis of cerebral malaria by artesunate may not only involve the killing of plasmodium but also the inhibition of a cytokine storm in the host. This study provides new insights into the molecular mechanism by which artesunate improves the prognosis of cerebral malaria.


Antimalarials , Artemisinins , Malaria, Cerebral , Animals , Anti-Inflammatory Agents/therapeutic use , Antimalarials/pharmacology , Antimalarials/therapeutic use , Artemisinins/pharmacology , Artemisinins/therapeutic use , Artesunate/pharmacology , Artesunate/therapeutic use , Disease Models, Animal , Gene Expression Profiling , Intercellular Adhesion Molecule-1/therapeutic use , Interleukin-10/therapeutic use , Interleukin-6/therapeutic use , Lead/therapeutic use , Malaria, Cerebral/drug therapy , Malaria, Cerebral/genetics , Malaria, Cerebral/metabolism , Mice , Mice, Inbred C57BL , RNA-Seq , Toll-Like Receptors/therapeutic use , Tumor Necrosis Factor-alpha/therapeutic use
5.
EMBO Rep ; 23(6): e53641, 2022 06 07.
Article En | MEDLINE | ID: mdl-35417070

Plasmodium falciparum, the deadliest form of human malaria, remains one of the major threats to human health in endemic regions. Its virulence is attributed to its ability to modify infected red blood cells (iRBC) to adhere to endothelial receptors by placing variable antigens known as PfEMP1 on the iRBC surface. PfEMP1 expression determines the cytoadhesive properties of the iRBCs and is implicated in severe malaria. To evade antibody-mediated responses, the parasite undergoes continuous switches of expression between different PfEMP1 variants. Recently, it became clear that in addition to antibody-mediated responses, PfEMP1 triggers innate immune responses; however, the role of neutrophils, the most abundant white blood cells in the human circulation, in malaria remains elusive. Here, we show that neutrophils recognize and kill blood-stage P. falciparum isolates. We identify neutrophil ICAM-1 and specific PfEMP1 implicated in cerebral malaria as the key molecules involved in this killing. Our data provide mechanistic insight into the interactions between neutrophils and iRBCs and demonstrate the important influence of PfEMP1 on the selective innate response to cerebral malaria.


Malaria, Cerebral , Malaria, Falciparum , Plasmodium falciparum , Erythrocytes/parasitology , Humans , Malaria, Cerebral/genetics , Malaria, Cerebral/metabolism , Malaria, Falciparum/genetics , Neutrophils/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/physiology , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
6.
J Infect Dis ; 225(10): 1786-1790, 2022 05 16.
Article En | MEDLINE | ID: mdl-34718631

Cerebral malaria (CM) may cause death or long-term neurological damage in children, and several host genetic risk factors have been reported. Malaria-specific immunoglobulin (Ig) G3 antibodies are crucial to human immune response against malaria. The hinge region of IgG3 exhibits length polymorphism (with long [L], medium [M], and short [S] alleles), which may influence its functionality. We studied IgG3 hinge region length polymorphisms in 136 Ghanaian children with malaria. Using logistic regression models, we found that children with the recessive MM allotype encoding medium IgG3 hinge region length had an increased risk of CM (adjusted odds ratio, 6.67 [95% confidence interval,1.30-34.32]; P=.004) . This has implications for future epidemiological studies on CM.


Antibodies, Protozoan , Immunoglobulin G , Malaria, Cerebral , Malaria, Falciparum , Antibodies, Protozoan/genetics , Child , Ghana/epidemiology , Humans , Immunoglobulin G/genetics , Malaria, Cerebral/epidemiology , Malaria, Cerebral/genetics , Malaria, Falciparum/epidemiology , Malaria, Falciparum/genetics , Plasmodium falciparum
7.
Front Cell Infect Microbiol ; 11: 743616, 2021.
Article En | MEDLINE | ID: mdl-34746025

Malaria is a major international public health problem that affects millions of patients worldwide especially in sub-Saharan Africa. Although many tests have been developed to diagnose malaria infections, we still lack reliable diagnostic biomarkers for the identification of disease severity, especially in endemic areas where the diagnosis of cerebral malaria is very difficult and requires the exclusion of all other possible causes. Previous host and pathogen transcriptomic studies have not yielded homogenous results that can be harnessed into a reliable diagnostic tool. Here we utilized a multi-cohort analysis approach using machine-learning algorithms to identify blood gene signatures that can distinguish severe and cerebral malaria from moderate and non-cerebral cases. Using a Regularized Random Forest model, we identified 28-gene and 32-gene signatures that can reliably distinguish severe and cerebral malaria, respectively. We tested the specificity of both signatures against other common infectious diseases to ensure the signatures reliability and suitability as diagnostic markers. The severe and cerebral malaria gene-signatures were further integrated through k-top scoring pairs classifiers into ten and nine gene pairs that could distinguish severe and cerebral malaria, respectively. These signatures have various implications that can be utilized as blood diagnostic tools for malaria severity in endemic countries.


Malaria, Cerebral , Malaria, Falciparum , Cohort Studies , Humans , Malaria, Cerebral/diagnosis , Malaria, Cerebral/genetics , Reproducibility of Results , Severity of Illness Index
8.
Malar J ; 20(1): 297, 2021 Jul 02.
Article En | MEDLINE | ID: mdl-34215257

BACKGROUND: Recent genome wide analysis studies have identified a strong association between single nucleotide variations within the human ATP2B4 gene and susceptibility to severe malaria. The ATP2B4 gene encodes the plasma membrane calcium ATPase 4 (PMCA4), which is responsible for controlling the physiological level of intracellular calcium in many cell types, including red blood cells (RBCs). It is, therefore, postulated that genetic differences in the activity or expression level of PMCA4 alters intracellular Ca2+ levels and affects RBC hydration, modulating the invasion and growth of the Plasmodium parasite within its target host cell. METHODS: In this study the course of three different Plasmodium spp. infections were examined in mice with systemic knockout of Pmca4 expression. RESULTS: Ablation of PMCA4 reduced the size of RBCs and their haemoglobin content but did not affect RBC maturation and reticulocyte count. Surprisingly, knockout of PMCA4 did not significantly alter peripheral parasite burdens or the dynamics of blood stage Plasmodium chabaudi infection or reticulocyte-restricted Plasmodium yoelii infection. Interestingly, although ablation of PMCA4 did not affect peripheral parasite levels during Plasmodium berghei infection, it did promote slight protection against experimental cerebral malaria, associated with a minor reduction in antigen-experienced T cell accumulation in the brain. CONCLUSIONS: The finding suggests that PMCA4 may play a minor role in the development of severe malarial complications, but that this appears independent of direct effects on parasite invasion, growth or survival within RBCs.


Disease Resistance/genetics , Malaria/genetics , Plasma Membrane Calcium-Transporting ATPases/genetics , Plasmodium/physiology , Animals , Cell Membrane , Malaria/blood , Malaria/parasitology , Malaria, Cerebral/genetics , Malaria, Cerebral/parasitology , Mice , Mice, Knockout , Plasma Membrane Calcium-Transporting ATPases/metabolism , Plasmodium berghei/physiology , Plasmodium chabaudi/physiology , Plasmodium yoelii/physiology
10.
Immun Inflamm Dis ; 9(3): 819-826, 2021 09.
Article En | MEDLINE | ID: mdl-33942992

INTRODUCTION: A vaccine for malaria is urgently required but no vaccine has yet shown satisfactory protective efficacy especially for Plasmodium falciparum. P. falciparum infection can progress to cerebral malaria (CM), a neurological syndrome with exceedingly high mortality. Designing effective P. falciparum vaccines require more understanding of the protective immune response while the host immune response to CM and the mechanisms are still elusive. Here, we aim to identify host gene responses to CM and host gene networks associated with CM pathogenesis. METHODS: An innovative genomic analysis strategy, the weighted gene coexpression network analysis (WGCNA) combined with differential gene expression analysis, was used in this study. Data for analysis contain 93 whole blood samples, derived from two previous public transcriptome datasets. RESULTS: This approach led to the identification of numerous differentially expressed human transcripts and dozens of coexpression gene modules. We further identified nine key genes, including MBP, SAMSN1, PSMF1, SLC39A8, EIF3B, SMPDL3A, FABP5, SPSB3, and SHARPIN, of which the last four genes were first identified to be related to CM in the present study. CONCLUSION: The results provided a comprehensive characterization of host gene expression profiles in CM and offered some new insight into malaria vaccine design. These identified key genes could be potential targets or immune modulators for novel therapeutic interventions of CM.


Malaria, Cerebral , Malaria, Falciparum , Adaptor Proteins, Vesicular Transport , Fatty Acid-Binding Proteins , Genomics , Humans , Immunity , Malaria, Cerebral/genetics , Malaria, Falciparum/genetics , Plasmodium falciparum/genetics
11.
J Biol Chem ; 296: 100615, 2021.
Article En | MEDLINE | ID: mdl-33798555

Persistent high levels of proinflammatory and Th1 responses contribute to cerebral malaria (CM). Suppression of inflammatory responses and promotion of Th2 responses prevent pathogenesis. IL-4 commonly promotes Th2 responses and inhibits inflammatory and Th1 responses. Therefore, IL-4 is widely considered as a beneficial cytokine via its Th2-promoting role that is predicted to provide protection against severe malaria by inhibiting inflammatory responses. However, IL-4 may also induce inflammatory responses, as the result of IL-4 action depends on the timing and levels of its production and the tissue environment in which it is produced. Recently, we showed that dendritic cells (DCs) produce IL-4 early during malaria infection in response to a parasite protein and that this IL-4 response may contribute to severe malaria. However, the mechanism by which IL-4 produced by DCs contributing to lethal malaria is unknown. Using Plasmodium berghei ANKA-infected C57BL/6 mice, a CM model, we show here that mice lacking IL-4Rα only in CD8α+ DCs are protected against CM pathogenesis and survive, whereas WT mice develop CM and die. Compared with WT mice, mice lacking IL-4Rα in CD11c+ or CD8α+ DCs showed reduced inflammatory responses leading to decreased Th1 and cytotoxic CD8+ T cell responses, lower infiltration of CD8+ T cells to the brain, and negligible brain pathology. The novel results presented here reveal a paradoxical role of IL-4Rα signaling in CM pathogenesis that promotes CD8α+ DC-mediated inflammatory responses that generate damaging Th1 and cytotoxic CD8+ T cell responses.


CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Receptors, Cell Surface/immunology , Signal Transduction/immunology , Th1 Cells/immunology , Animals , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/pathology , Dendritic Cells/pathology , Interleukin-4/genetics , Interleukin-4/immunology , Malaria, Cerebral/genetics , Malaria, Cerebral/pathology , Mice , Mice, Knockout , Plasmodium berghei/genetics , Receptors, Cell Surface/genetics , Signal Transduction/genetics , Th1 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology
12.
Int J Biol Macromol ; 177: 535-547, 2021 Apr 30.
Article En | MEDLINE | ID: mdl-33631268

Cerebral malaria (CM) is the most severe complication caused by Plasmodium falciparum infection. The pathophysiological changes caused by parasite virulence factors and the human immune response to parasites contribute to CM. To date, very few parasite virulence proteins have been found to participate in CM. Here, we employed comparative genomics analysis and identified parasite-infected erythrocyte specific protein 2 (PIESP2) to be a CM-related protein. We conducted further experimental investigations and found that PIESP2 is an immunogenic protein. PIESP2 expression begins at the early trophozoite stage and progressively increases with parasite development. Although PIESP2 proteins mainly reside within infected red blood cells (IRBCs), some of them are present on the IRBC surface at the pigmented stage. Moreover, blockage of PIESP2 by antiserum apparently inhibited the adhesion of IRBCs to brain microvascular endothelial cells (BMECs). Western blot analysis detected the binding of PIESP2 to BMECs. Transcriptional analysis revealed that the binding of PIESP2 to BMECs can increase the expression of genes involved in the inflammatory response but decrease the expression of genes related to the anchoring junction. Overall, PIESP2 might be associated with CM by mediating the sequestration of IRBCs, inducing the inflammation response, and impairing the integrity of blood-brain barrier.


Malaria, Cerebral/genetics , Malaria, Falciparum/genetics , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Virulence Factors/genetics , Humans , Malaria, Cerebral/metabolism , Malaria, Falciparum/metabolism , Plasmodium falciparum/metabolism , Plasmodium falciparum/pathogenicity , Protozoan Proteins/metabolism , Virulence Factors/metabolism
13.
Sci Rep ; 11(1): 3680, 2021 02 11.
Article En | MEDLINE | ID: mdl-33574457

The Plasmodium falciparum erythrocyte-membrane-protein-1 (PF3D7_1150400/PF11_0521) contains both domain cassette DC13 and DBLß3 domain binding to EPCR and ICAM-1 receptors, respectively. This type of PfEMP1 proteins with dual binding specificity mediate specific interactions with brain micro-vessels endothelium leading to the development of cerebral malaria (CM). Using plasma collected from children at time of hospital admission and after 30 days, we study an acquisition of IgG response to PF3D7_1150400/PF11_0521 DC13 and DBLß3_D4 recombinant constructs, and five peptides located within these constructs, specifically in DBLα1.7_D2 and DBLß3_D4 domains. We found significant IgG responses against the entire DC13, PF11_0521_DBLß3_D4 domain, and peptides. The responses varied against different peptides and depended on the clinical status of children. The response was stronger at day 30, and mostly did not differ between CM and uncomplicated malaria (UM) groups. Specifically, the DBLß3 B3-34 peptide that contains essential residues involved in the interaction between PF11_0521 DBLß3_D4 domain and ICAM-1 receptor demonstrated significant increase in reactivity to IgG1 and IgG3 antibodies at convalescence. Further, IgG reactivity in CM group at time of admission against functionally active (ICAM-1-binding) PF11_0521 DBLß3_D4 domain was associated with protection against severe anemia. These results support development of vaccine based on the PF3D7_1150400/PF11_0521 structures to prevent CM.


Immunoglobulin G/blood , Malaria, Cerebral/immunology , Malaria, Falciparum/immunology , Peptides/immunology , Protozoan Proteins/immunology , Anemia/complications , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Antigens, Protozoan/blood , Antigens, Protozoan/immunology , Brain/immunology , Brain/metabolism , Brain/parasitology , Brain/pathology , Child, Preschool , Endothelial Protein C Receptor/genetics , Endothelial Protein C Receptor/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/parasitology , Erythrocytes/parasitology , Female , Humans , Immunoglobulin G/immunology , Infant , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Malaria, Cerebral/blood , Malaria, Cerebral/genetics , Malaria, Cerebral/parasitology , Malaria, Falciparum/blood , Malaria, Falciparum/genetics , Malaria, Falciparum/parasitology , Male , Peptides/genetics , Plasmodium falciparum/genetics , Plasmodium falciparum/pathogenicity , Protein Binding/genetics , Protein Binding/immunology , Protozoan Proteins/genetics
14.
Nat Commun ; 11(1): 4596, 2020 09 14.
Article En | MEDLINE | ID: mdl-32929083

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.


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
15.
Mediators Inflamm ; 2020: 3280689, 2020.
Article En | MEDLINE | ID: mdl-32801995

BACKGROUND: Cerebral malaria (CM), a reversible encephalopathy affecting young children, is a medical emergency requiring rapid clinical assessment and treatment. However, understanding of the genes/proteins and the biological pathways involved in the disease outcome is still limited. METHODS: We have performed a whole transcriptomic analysis of blood samples from Malian children with CM or uncomplicated malaria (UM). Hierarchical clustering and pathway, network, and upstream regulator analyses were performed to explore differentially expressed genes (DEGs). We validated gene expression for 8 genes using real-time quantitative PCR (RT-qPCR). Plasma levels were measured for IP-10/CXCL10 and IL-18. RESULTS: A blood RNA signature including 538 DEGs (∣FC | ≥2.0, adjusted P value ≤ 0.01) allowed to discriminate between CM and UM. Ingenuity Pathway Analysis (IPA) and Kyoto Encyclopedia of Genes and Genomes (KEGG) revealed novel genes and biological pathways related to immune/inflammatory responses, erythrocyte alteration, and neurodegenerative disorders. Gene expressions of CXCL10, IL12RB2, IL18BP, IL2RA, AXIN2, and NET were significantly lower in CM whereas ARG1 and SLC6A9 were higher in CM compared to UM. Plasma protein levels of IP-10/CXCL10 were significantly lower in CM than in UM while levels of IL-18 were higher. Interestingly, among children with CM, those who died from a complication of malaria tended to have higher concentrations of IP-10/CXCL10 and IFN-γ than those who recovered. CONCLUSIONS: This study identified some new factors and mechanisms that play crucial roles in CM and characterized their respective biological pathways as well as some upstream regulators.


Brain/metabolism , Erythrocytes/metabolism , Inflammation/blood , Malaria, Cerebral/genetics , Malaria, Cerebral/metabolism , Transcriptome/genetics , Chemokine CXCL10/blood , Computational Biology/methods , Humans , Interleukin-18/blood , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
16.
J Leukoc Biol ; 108(4): 1183-1197, 2020 10.
Article En | MEDLINE | ID: mdl-32362022

Heterogeneity and high plasticity are common features of cells from the mononuclear phagocyte system: monocytes (MOs), macrophages, and dendritic cells (DCs). Upon activation by microbial agents, MO can differentiate into MO-derived DCs (MODCs). In previous work, we have shown that during acute infection with Plasmodium berghei ANKA (PbA), MODCs become, transiently, the main CD11b+ myeloid population in the spleen (SP) and once recruited to the brain play an important role in the development of experimental cerebral malaria (ECM). Here, we isolated 4 cell populations: bone marrow (BM) MOs (BM-MOs) and SP-MOs from uninfected mice; BM inflammatory MOs (BM-iMOs) and SP-MODCs from PbA-infected mice and used a system biology approach to a holistic transcriptomic comparison and provide an interactome analysis by integrating differentially expressed miRNAs (DEMs) and their differentially expressed gene targets (DEGs) data. The Jaccard index (JI) was used for gauging the similarity and diversity among these cell populations. Whereas BM-MOs, BM-iMOs, and SP-MOs presented high similarity of DEGs, SP-MODCs distinguished by showing a greater number of DEGs. Moreover, functional analysis identified an enrichment in canonical pathways, such as DC maturation, neuroinflammation, and IFN signaling. Upstream regulator analysis identified IFNγ as the potential upstream molecule that can explain the observed DEMs-Target DEGs intersections in SP-MODCs. Finally, directed target analysis and in vivo/ex vivo assays indicate that SP-MODCs differentiate in the SP and IFNγ is a main driver of this process.


Dendritic Cells/immunology , Gene Expression Regulation/immunology , Malaria, Cerebral/immunology , MicroRNAs/immunology , Monocytes/immunology , Plasmodium berghei/immunology , RNA, Messenger/immunology , Animals , Dendritic Cells/pathology , Malaria, Cerebral/genetics , Malaria, Cerebral/pathology , Mice , Mice, Knockout , MicroRNAs/genetics , Monocytes/pathology , RNA, Messenger/genetics , Transcriptome/immunology
17.
PLoS Pathog ; 16(1): e1008261, 2020 01.
Article En | MEDLINE | ID: mdl-31999807

Disruption of blood-brain barrier (BBB) function is a key feature of cerebral malaria. Increased barrier permeability occurs due to disassembly of tight and adherens junctions between endothelial cells, yet the mechanisms governing junction disassembly and vascular permeability during cerebral malaria remain poorly characterized. We found that EphA2 is a principal receptor tyrosine kinase mediating BBB breakdown during Plasmodium infection. Upregulated on brain microvascular endothelial cells in response to inflammatory cytokines, EphA2 is required for the loss of junction proteins on mouse and human brain microvascular endothelial cells. Furthermore, EphA2 is necessary for CD8+ T cell brain infiltration and subsequent BBB breakdown in a mouse model of cerebral malaria. Blocking EphA2 protects against BBB breakdown highlighting EphA2 as a potential therapeutic target for cerebral malaria.


Blood-Brain Barrier/parasitology , Malaria, Cerebral/parasitology , Receptor, EphA2/metabolism , Adolescent , Animals , Blood-Brain Barrier/metabolism , Child , Child, Preschool , Cross-Sectional Studies , Female , Humans , Infant , Malaria, Cerebral/genetics , Malaria, Cerebral/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasmodium falciparum/physiology , Receptor, EphA2/genetics
18.
Genes Immun ; 21(1): 45-62, 2020 01.
Article En | MEDLINE | ID: mdl-31501529

Genetic mapping and genome-wide studies provide evidence for the association of several genetic polymorphisms with malaria, a complex pathological disease with multiple severity degrees. We have previously described Berr1and Berr2 as candidate genes identified in the WLA/Pas inbreed mouse strain predisposing to resistance to cerebral malaria (CM) induced by P. berghei ANKA. We report in this study the phenotypic and functional characteristics of a congenic strain we have derived for Berr2WLA allele on the C57BL/6JR (B6) background. B6.WLA-Berr2 was found highly resistant to CM compared to C57BL/6JR susceptible mice. The mechanisms associated with CM resistance were analyzed by combining genotype, transcriptomic and immune response studies. We found that B6.WLA-Berr2 mice showed a reduced parasite sequestration and blood-brain barrier disruption with low CXCR3+ T cell infiltration in the brain along with altered glial cell response upon P. berghei ANKA infection compared to B6. In addition, we have identified the CD300f, belonging to a family of Ig-like encoding genes, as a potential candidate associated with CM resistance. Microglia cells isolated from the brain of infected B6.WLA-Berr2 mice significantly expressed higher level of CD300f compared to CMS mice and were associated with inhibition of inflammatory response.


Malaria, Cerebral/genetics , Microglia/metabolism , Receptors, Immunologic/metabolism , Alleles , Animals , Brain/immunology , Brain/metabolism , Brain/pathology , Chromosome Mapping , Disease Models, Animal , Disease Resistance/genetics , Disease Resistance/immunology , Female , Genotype , Malaria, Cerebral/metabolism , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Microglia/physiology , Receptors, Immunologic/genetics
19.
Infect Immun ; 88(2)2020 01 22.
Article En | MEDLINE | ID: mdl-31792077

We used a genome-wide screen in N-ethyl-N-nitrosourea (ENU)-mutagenized mice to identify genes in which recessive loss-of-function mutations protect against pathological neuroinflammation. We identified an R367Q mutation in the ZBTB7B (ThPOK) protein in which homozygosity causes protection against experimental cerebral malaria (ECM) caused by infection with Plasmodium berghei ANKA. Zbtb7bR367Q homozygous mice show a defect in the lymphoid compartment expressed as severe reduction in the number of single-positive CD4 T cells in the thymus and in the periphery, reduced brain infiltration of proinflammatory leukocytes in P. berghei ANKA-infected mice, and reduced production of proinflammatory cytokines by primary T cells ex vivo and in vivo Dampening of proinflammatory immune responses in Zbtb7bR367Q mice is concomitant to increased susceptibility to infection with avirulent (Mycobacterium bovis BCG) and virulent (Mycobacterium tuberculosis H37Rv) mycobacteria. The R367Q mutation maps to the first DNA-binding zinc finger domain of ThPOK and causes loss of base contact by R367 in the major groove of the DNA, which is predicted to impair DNA binding. Global immunoprecipitation of ThPOK-containing chromatin complexes coupled to DNA sequencing (ChIP-seq) identified transcriptional networks and candidate genes likely to play key roles in CD4+ CD8+ T cell development and in the expression of lineage-specific functions of these cells. This study highlights ThPOK as a global regulator of immune function in which alterations may affect normal responses to infectious and inflammatory stimuli.


DNA-Binding Proteins/genetics , Malaria, Cerebral/genetics , Transcription Factors/genetics , Tuberculosis, Pulmonary/genetics , Animals , Brain/microbiology , CD4-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/microbiology , Cytokines/genetics , Female , Inflammation/genetics , Inflammation/microbiology , Malaria, Cerebral/microbiology , Male , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/pathogenicity , Plasmodium berghei/pathogenicity , Tuberculosis, Pulmonary/microbiology , Virulence/genetics
20.
BMC Med Genomics ; 12(1): 148, 2019 10 30.
Article En | MEDLINE | ID: mdl-31666081

BACKGROUND: Plasmodium falciparum malaria remains a major health problem in Africa. The mechanisms of pathogenesis are not fully understood. Transcriptomic studies may provide new insights into molecular pathways involved in the severe form of the disease. METHODS: Blood transcriptional levels were assessed in patients with cerebral malaria, non-cerebral malaria, or mild malaria by using microarray technology to look for gene expression profiles associated with clinical status. Multi-way ANOVA was used to extract differentially expressed genes. Network and pathways analyses were used to detect enrichment for biological pathways. RESULTS: We identified a set of 443 genes that were differentially expressed in the three patient groups after applying a false discovery rate of 10%. Since the cerebral patients displayed a particular transcriptional pattern, we focused our analysis on the differences between cerebral malaria patients and mild malaria patients. We further found 842 differentially expressed genes after applying a false discovery rate of 10%. Unsupervised hierarchical clustering of cerebral malaria-informative genes led to clustering of the cerebral malaria patients. The support vector machine method allowed us to correctly classify five out of six cerebral malaria patients and six of six mild malaria patients. Furthermore, the products of the differentially expressed genes were mapped onto a human protein-protein network. This led to the identification of the proteins with the highest number of interactions, including GSK3B, RELA, and APP. The enrichment analysis of the gene functional annotation indicates that genes involved in immune signalling pathways play a role in the occurrence of cerebral malaria. These include BCR-, TCR-, TLR-, cytokine-, FcεRI-, and FCGR- signalling pathways and natural killer cell cytotoxicity pathways, which are involved in the activation of immune cells. In addition, our results revealed an enrichment of genes involved in Alzheimer's disease. CONCLUSIONS: In the present study, we examine a set of genes whose expression differed in cerebral malaria patients and mild malaria patients. Moreover, our results provide new insights into the potential effect of the dysregulation of gene expression in immune pathways. Host genetic variation may partly explain such alteration of gene expression. Further studies are required to investigate this in African populations.


Malaria, Cerebral/pathology , Transcriptome/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Cluster Analysis , Female , Glycogen Synthase Kinase 3 beta/genetics , Humans , Infant , Malaria, Cerebral/blood , Malaria, Cerebral/genetics , Male , Middle Aged , Protein Interaction Maps/genetics , Senegal , Severity of Illness Index , Signal Transduction , Transcription Factor RelA/genetics , Young Adult
...