Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 230
Filter
1.
J Neuroinflammation ; 21(1): 119, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715061

ABSTRACT

BACKGROUND: Cerebral malaria (CM) is the most lethal complication of malaria, and survivors usually endure neurological sequelae. Notably, the cytotoxic effect of infiltrating Plasmodium-activated CD8+ T cells on cerebral microvasculature endothelial cells is a prominent feature of the experimental CM (ECM) model with blood-brain barrier disruption. However, the damage effect of CD8+ T cells infiltrating the brain parenchyma on neurons remains unclear. Based on the immunosuppressive effect of the PD-1/PD-L1 pathway on T cells, our previous study demonstrated that the systemic upregulation of PD-L1 to inhibit CD8+ T cell function could effectively alleviate the symptoms of ECM mice. However, it has not been reported whether neurons can suppress the pathogenic effect of CD8+ T cells through the PD-1/PD-L1 negative immunomodulatory pathway. As the important inflammatory factor of CM, interferons can induce the expression of PD-L1 via different molecular mechanisms according to the neuro-immune microenvironment. Therefore, this study aimed to investigate the direct interaction between CD8+ T cells and neurons, as well as the mechanism of neurons to alleviate the pathogenic effect of CD8+ T cells through up-regulating PD-L1 induced by IFNs. METHODS: Using the ECM model of C57BL/6J mice infected with Plasmodium berghei ANKA (PbA), morphological observations were conducted in vivo by electron microscope and IF staining. The interaction between the ECM CD8+ T cells (immune magnetic bead sorting from spleen of ECM mice) and primary cultured cortical neurons in vitro was observed by IF staining and time-lapse photography. RNA-seq was performed to analyze the signaling pathway of PD-L1 upregulation in neurons induced by IFNß or IFNγ, and verified through q-PCR, WB, IF staining, and flow cytometry both in vitro and in vivo using IFNAR or IFNGR gene knockout mice. The protective effect of adenovirus-mediated PD-L1 IgGFc fusion protein expression was verified in ECM mice with brain stereotaxic injection in vivo and in primary cultured neurons via viral infection in vitro. RESULTS: In vivo, ECM mice showed infiltration of activated CD8+ T cells and neuronal injury in the brain parenchyma. In vitro, ECM CD8+ T cells were in direct contact with neurons and induced axonal damage, as an active behavior. The PD-L1 protein level was elevated in neurons of ECM mice and in primary cultured neurons induced by IFNß, IFNγ, or ECM CD8+ T cells in vitro. Furthermore, the IFNß or IFNγ induced neuronal expression of PD-L1 was mediated by increasing STAT1/IRF1 pathway via IFN receptors. The increase of PD-L1 expression in neurons during PbA infection was weakened after deleting the IFNAR or IFNGR. Increased PD-L1 expression by adenovirus partially protected neurons from CD8+ T cell-mediated damage both in vitro and in vivo. CONCLUSION: Our study demonstrates that both type I and type II IFNs can induce neurons to upregulate PD-L1 via the STAT1/IRF1 pathway mediated by IFN receptors to protect against activated CD8+ T cell-mediated damage, providing a targeted pathway to alleviate neuroinflammation during ECM.


Subject(s)
B7-H1 Antigen , CD8-Positive T-Lymphocytes , Malaria, Cerebral , Mice, Inbred C57BL , Neurons , STAT1 Transcription Factor , Up-Regulation , Animals , Mice , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Interferon Regulatory Factor-1/metabolism , Interferon-gamma/metabolism , Malaria, Cerebral/immunology , Malaria, Cerebral/metabolism , Malaria, Cerebral/pathology , Mice, Knockout , Neurons/metabolism , Plasmodium berghei , Signal Transduction/physiology , STAT1 Transcription Factor/metabolism , Up-Regulation/drug effects
2.
Fluids Barriers CNS ; 21(1): 38, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693577

ABSTRACT

BACKGROUND: Blood-brain barrier (BBB) disruption is a central feature of cerebral malaria (CM), a severe complication of Plasmodium falciparum (Pf) infections. In CM, sequestration of Pf-infected red blood cells (Pf-iRBCs) to brain endothelial cells combined with inflammation, hemolysis, microvasculature obstruction and endothelial dysfunction mediates BBB disruption, resulting in severe neurologic symptoms including coma and seizures, potentially leading to death or long-term sequelae. In vitro models have advanced our knowledge of CM-mediated BBB disruption, but their physiological relevance remains uncertain. Using human induced pluripotent stem cell-derived brain microvascular endothelial cells (hiPSC-BMECs), we aimed to develop a novel in vitro model of the BBB in CM, exhibiting enhanced barrier properties. METHODS: hiPSC-BMECs were co-cultured with HB3var03 strain Pf-iRBCs up to 9 h. Barrier integrity was measured using transendothelial electrical resistance (TEER) and sodium fluorescein permeability assays. Localization and expression of tight junction (TJ) proteins (occludin, zonula occludens-1, claudin-5), cellular adhesion molecules (ICAM-1, VCAM-1), and endothelial surface markers (EPCR) were determined using immunofluorescence imaging (IF) and western blotting (WB). Expression of angiogenic and cell stress markers were measured using multiplex proteome profiler arrays. RESULTS: After 6-h of co-culture with Pf-iRBCs, hiPSC-BMECs showed reduced TEER and increased sodium fluorescein permeability compared to co-culture with uninfected RBCs, indicative of a leaky barrier. We observed disruptions in localization of occludin, zonula occludens-1, and claudin-5 by IF, but no change in protein expression by WB in Pf-iRBC co-cultures. Expression of ICAM-1 and VCAM-1 but not EPCR was elevated in hiPSC-BMECs with Pf-iRBC co-culture compared to uninfected RBC co-culture. In addition, there was an increase in expression of angiogenin, platelet factor-4, and phospho-heat shock protein-27 in the Pf-iRBCs co-culture compared to uninfected RBC co-culture. CONCLUSION: These findings demonstrate the validity of our hiPSC-BMECs based model of the BBB, that displays enhanced barrier integrity and appropriate TJ protein localization. In the hiPSC-BMEC co-culture with Pf-iRBCs, reduced TEER, increased paracellular permeability, changes in TJ protein localization, increase in expression of adhesion molecules, and markers of angiogenesis and cellular stress all point towards a novel model with enhanced barrier properties, suitable for investigating pathogenic mechanisms underlying BBB disruption in CM.


Subject(s)
Blood-Brain Barrier , Induced Pluripotent Stem Cells , Malaria, Cerebral , Blood-Brain Barrier/metabolism , Humans , Malaria, Cerebral/metabolism , Endothelial Cells/metabolism , Cells, Cultured , Coculture Techniques , Models, Biological
3.
Brain Res ; 1822: 148669, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37951562

ABSTRACT

Cerebral malaria (CM) pathogenesis is described as a multistep mechanism. In this context, monocytes have been implicated in CM pathogenesis by increasing the sequestration of infected red blood cells to the brain microvasculature. In disease, endothelial activation is followed by reduced monocyte rolling and increased adhesion. Nowadays, an important challenge is to identify potential pro-inflammatory stimuli that can modulate monocytes behavior. Our group have demonstrated that bradykinin (BK), a pro-inflammatory peptide involved in CM, is generated during the erythrocytic cycle of P. falciparum and is detected in culture supernatant (conditioned medium). Herein we investigated the role of BK in the adhesion of monocytes to endothelial cells of blood brain barrier (BBB). To address this issue human monocytic cell line (THP-1) and human brain microvascular endothelial cells (hBMECs) were used. It was observed that 20% conditioned medium from P. falciparum infected erythrocytes (Pf-iRBC sup) increased the adhesion of THP-1 cells to hBMECs. This effect was mediated by BK through the activation of B2 and B1 receptors and involves the increase in ICAM-1 expression in THP-1 cells. Additionally, it was observed that angiotensin-converting enzyme (ACE) inhibitor, captopril, enhanced the effect of both BK and Pf-iRBC sup on THP-1 adhesion. Together these data show that BK, generated during the erythrocytic cycle of P. falciparum, could play an important role in adhesion of monocytes in endothelial cells lining the BBB.


Subject(s)
Blood-Brain Barrier , Bradykinin , Cell Adhesion , Malaria, Cerebral , Malaria, Falciparum , Plasmodium falciparum , Humans , Bradykinin/metabolism , Cell Adhesion/physiology , Culture Media, Conditioned/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Erythrocytes/parasitology , Malaria, Cerebral/metabolism , Malaria, Cerebral/parasitology , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Monocytes/physiology , Plasmodium falciparum/physiology , Blood-Brain Barrier/physiopathology
4.
Mem Inst Oswaldo Cruz ; 118: e230033, 2023.
Article in English | MEDLINE | ID: mdl-37403869

ABSTRACT

BACKGROUND: Cerebral malaria (CM) is a severe immunovasculopathy caused for Plasmodium falciparum infection, which is characterised by the sequestration of parasitised red blood cells (pRBCs) in brain microvessels. Previous studies have shown that some terpenes, such as perillyl alcohol (POH), exhibit a marked efficacy in preventing cerebrovascular inflammation, breakdown of the brain-blood barrier (BBB) and brain leucocyte accumulation in experimental CM models. OBJECTIVE: To analyse the effects of POH on the endothelium using human brain endothelial cell (HBEC) monolayers co-cultured with pRBCs. METHODOLOGY: The loss of tight junction proteins (TJPs) and features of endothelial activation, such as ICAM-1 and VCAM-1 expression were evaluated by quantitative immunofluorescence. Microvesicle (MV) release by HBEC upon stimulation by P. falciparum was evaluated by flow cytometry. Finally, the capacity of POH to revert P. falciparum-induced HBEC monolayer permeability was examined by monitoring trans-endothelial electrical resistance (TEER). FINDINGS: POH significantly prevented pRBCs-induced endothelial adhesion molecule (ICAM-1, VCAM-1) upregulation and MV release by HBEC, improved their trans-endothelial resistance, and restored their distribution of TJPs such as VE-cadherin, Occludin, and JAM-A. CONCLUSIONS: POH is a potent monoterpene that is efficient in preventing P. falciparum-pRBCs-induced changes in HBEC, namely their activation, increased permeability and alterations of integrity, all parameters of relevance to CM pathogenesis.


Subject(s)
Malaria, Cerebral , Malaria, Falciparum , Humans , Plasmodium falciparum , Intercellular Adhesion Molecule-1/metabolism , Endothelial Cells , Vascular Cell Adhesion Molecule-1/metabolism , Brain/metabolism , Brain/pathology , Malaria, Cerebral/metabolism , Malaria, Cerebral/pathology , Monoterpenes/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Endothelium, Vascular , Permeability
5.
Cells ; 12(7)2023 03 23.
Article in English | MEDLINE | ID: mdl-37048057

ABSTRACT

Neural injuries in cerebral malaria patients are a significant cause of morbidity and mortality. Nevertheless, a comprehensive research approach to study this issue is lacking, so herein we propose an in vitro system to study human cerebral malaria using cellular approaches. Our first goal was to establish a cellular system to identify the molecular alterations in human brain vasculature cells that resemble the blood-brain barrier (BBB) in cerebral malaria (CM). Through transcriptomic analysis, we characterized specific gene expression profiles in human brain microvascular endothelial cells (HBMEC) activated by the Plasmodium falciparum parasites. We also suggest potential new genes related to parasitic activation. Then, we studied its impact at brain level after Plasmodium falciparum endothelial activation to gain a deeper understanding of the physiological mechanisms underlying CM. For that, the impact of HBMEC-P. falciparum-activated secretomes was evaluated in human brain organoids. Our results support the reliability of in vitro cellular models developed to mimic CM in several aspects. These systems can be of extreme importance to investigate the factors (parasitological and host) influencing CM, contributing to a molecular understanding of pathogenesis, brain injury, and dysfunction.


Subject(s)
Malaria, Cerebral , Humans , Malaria, Cerebral/metabolism , Malaria, Cerebral/parasitology , Malaria, Cerebral/pathology , Endothelial Cells/metabolism , Reproducibility of Results , Brain/pathology , Plasmodium falciparum , Organoids/metabolism
6.
PLoS One ; 18(1): e0280282, 2023.
Article in English | MEDLINE | ID: mdl-36626386

ABSTRACT

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.


Subject(s)
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
7.
Parasitol Res ; 122(3): 729-737, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36694092

ABSTRACT

Cerebral malaria (CM) is a severe neurological condition caused by Plasmodium falciparum. Disruption of the brain-blood barrier (BBB) is a key pathological event leading to brain edema and vascular leakage in both humans and in the mouse model of CM. Interactions of brain endothelial cells with infected red blood cells (iRBCs) and with circulating inflammatory mediators and immune cells contribute to BBB dysfunction in CM. Adjunctive therapies for CM aim at preserving the BBB to prevent neurologic deficits. Experimental animal and cellular models are essential to develop new therapeutic strategies. However, in mice, the disease develops rapidly, which offers a very narrow time window for testing the therapeutic potential of drugs acting in the BBB. Here, we establish a brain endothelial cell barrier whose disturbance can be monitored by several parameters. Using this system, we found that incubation with iRBCs and with extracellular particles (EPs) released by iRBCs changes endothelial cell morphology, decreases the tight junction protein zonula occludens-1 (ZO-1), increases the gene expression of the intercellular adhesion molecule 1 (ICAM-1), and induces a significant reduction in transendothelial electrical resistance (TEER) with increased permeability. We propose this in vitro experimental setup as a straightforward tool to investigate molecular interactions and pathways causing endothelial barrier dysfunction and to test compounds that may target BBB and be effective against CM. A pre-selection of the effective compounds that strengthen the resistance of the brain endothelial cell barrier to Plasmodium-induced blood factors in vitro may increase the likelihood of their efficacy in preclinical disease mouse models of CM and in subsequent clinical trials with patients.


Subject(s)
Endothelial Cells , Malaria, Cerebral , Humans , Animals , Mice , Brain/metabolism , Blood-Brain Barrier , Malaria, Cerebral/drug therapy , Malaria, Cerebral/metabolism , Plasmodium falciparum/physiology
8.
Mol Omics ; 18(8): 716-730, 2022 09 26.
Article in English | MEDLINE | ID: mdl-35960011

ABSTRACT

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.


Subject(s)
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
9.
Mol Brain ; 15(1): 57, 2022 06 20.
Article in English | MEDLINE | ID: mdl-35725567

ABSTRACT

Cerebral malaria is the most serious complication of malaria infection, with 26% of surviving children having neurological sequelae, which may be caused by neuron damage, but the mechanism is not clear. Ferroptosis has been reported to play an important role in neuron damage in several nervous system diseases. However, the occurrence of ferroptosis in experimental cerebral malaria (ECM) pathogenesis is still unknown. In this study, we firstly detected increased levels of malondialdehyde (MDA) and iron, which are indicators of ferroptosis, in the cerebrum of ECM mice. Some important regulators of ferroptosis, including upregulated expression of transferrin receptor 1 (TfR1) and acyl-CoA synthetase long-chain family member 4 (ACSL4), and downregulation of glutathione peroxidase 4 (GPX4) levels, were also confirmed in ECM mice. Consistently, neuron damage, which was detected in the cerebrum of ECM mice, was positively correlated with reduced GPX4 expression and furtherly rescued by administration of the ferroptosis inhibitor ferrostatin-1 (Fer-1). In addition, primary neurons were damaged by activated CD8+ T cells, an effect that was also partially rescued by Fer-1 on amyloid precursor protein expression and mitochondrial membrane potential levels in vitro. Activated CD8+ T cells were also shown to infiltrate the cerebrum of ECM mice and upregulate TfR1 expression in primary neurons, which may be an important event for inducing ferroptosis in ECM. Altogether, we show that ferroptosis contributes to neuron damage in ECM pathogenesis, and activated CD8+ T cells may be important inducers of neuronal ferroptosis. Hence, targeting ferroptosis may be a promising adjuvant therapeutic strategy for neurological sequelae in patients with cerebral malaria.


Subject(s)
Ferroptosis , Malaria, Cerebral , Animals , CD8-Positive T-Lymphocytes , Malaria, Cerebral/metabolism , Malaria, Cerebral/pathology , Mice , Neurons/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase
10.
J Infect Dis ; 226(8): 1470-1479, 2022 10 17.
Article in English | MEDLINE | ID: mdl-35556124

ABSTRACT

BACKGROUND: Cerebral malaria (CM) is a rare, but severe and frequently fatal outcome of infection with Plasmodium falciparum. Pathogenetic mechanisms include endothelial activation and sequestration of parasitized erythrocytes in the cerebral microvessels. Increased concentrations of glycosaminoglycans in urine and plasma of malaria patients have been described, suggesting involvement of endothelial glycocalyx. METHODS: We used lectin histochemistry on postmortem samples to compare the distribution of multiple sugar epitopes on cerebral capillaries in children who died from CM and from nonmalarial comas. RESULTS: N-acetyl glucosamine residues detected by tomato lectin are generally reduced in children with CM compared to controls. We used the vascular expression of intercellular adhesion molecule 1 and mannose residues on brain capillaries of CM as evidence of local vascular inflammation, and both were expressed more highly in CM patients than controls. Sialic acid residues were found to be significantly reduced in patients with CM. By contrast, the levels of other sugar epitopes regularly detected on the cerebral vasculature were unchanged, and this suggests specific remodeling of cerebral microvessels in CM patients. CONCLUSIONS: Our findings support and expand upon earlier reports of disruptions of the endothelial glycocalyx in children with severe malaria.


Subject(s)
Malaria, Cerebral , Malaria, Falciparum , Brain/pathology , Capillaries/pathology , Child , Epitopes/metabolism , Erythrocytes/metabolism , Glucosamine/metabolism , Glycocalyx/metabolism , Glycosaminoglycans/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Lectins , Malaria, Cerebral/metabolism , Mannose/metabolism , N-Acetylneuraminic Acid/metabolism , Plasmodium falciparum/physiology
11.
EMBO Rep ; 23(6): e53641, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35417070

ABSTRACT

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.


Subject(s)
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
12.
Sci Adv ; 8(14): eabj7110, 2022 04 08.
Article in English | MEDLINE | ID: mdl-35385300

ABSTRACT

The modulation of the host's metabolism to protect tissue from damage induces tolerance to infections increasing survival. Here, we examined the role of the thyroid hormones, key metabolic regulators, in the outcome of malaria. Hypothyroidism confers protection to experimental cerebral malaria by a disease tolerance mechanism. Hypothyroid mice display increased survival after infection with Plasmodium berghei ANKA, diminishing intracranial pressure and brain damage, without altering pathogen burden, blood-brain barrier disruption, or immune cell infiltration. This protection is reversed by treatment with a Sirtuin 1 inhibitor, while treatment of euthyroid mice with a Sirtuin 1 activator induces tolerance and reduces intracranial pressure and lethality. This indicates that thyroid hormones and Sirtuin 1 are previously unknown targets for cerebral malaria treatment, a major killer of children in endemic malaria areas.


Subject(s)
Hypothyroidism , Malaria, Cerebral , Sirtuin 1 , Animals , Brain/metabolism , Disease Models, Animal , Hypothyroidism/metabolism , Malaria, Cerebral/drug therapy , Malaria, Cerebral/metabolism , Mice , Mice, Inbred C57BL , Plasmodium berghei , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/metabolism
13.
J Infect Dis ; 225(12): 2187-2196, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35255125

ABSTRACT

Cerebral malaria (CM) is the severest form of Plasmodium falciparum infection. Children under 5 years old are those most vulnerable to CM, and they consequently have the highest risk of malaria-related death. Parasite-associated factors leading to CM are not yet fully elucidated. We therefore sought to characterize the gene expression profile associated with CM, using RNA sequencing data from 15 CM and 15 uncomplicated malaria isolates from Benin. Cerebral malaria parasites displayed reduced circulation times, possibly related to higher cytoadherence capacity. Consistent with the latter, we detected increased var genes abundance in CM isolates. Differential expression analyses showed that distinct transcriptome profiles are signatures of malaria severity. Genes involved in adhesion, excluding variant surface antigens, were dysregulated, supporting the idea of increased cytoadhesion capacity of CM parasites. Finally, we found dysregulated expression of genes in the entry into host pathway that may reflect greater erythrocyte invasion capacity of CM parasites.


Subject(s)
Malaria, Cerebral , Malaria, Falciparum , Benin , Child , Child, Preschool , Erythrocytes/parasitology , Gene Expression Profiling , Humans , Malaria, Cerebral/metabolism , Malaria, Falciparum/metabolism , Plasmodium falciparum , Protozoan Proteins/metabolism , Transcriptome
14.
Mol Neurobiol ; 59(2): 800-820, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34782951

ABSTRACT

Long-term cognitive impairment associated with seizure-induced hippocampal damage is the key feature of cerebral malaria (CM) pathogenesis. One-fourth of child survivors of CM suffer from long-lasting neurological deficits and behavioral anomalies. However, mechanisms on hippocampal dysfunction are unclear. In this study, we elucidated whether gp91phox isoform of nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2) (a potent marker of oxidative stress) mediates hippocampal neuronal abnormalities and cognitive dysfunction in experimental CM (ECM). Mice symptomatic to CM were rescue treated with artemether monotherapy (ARM) and in combination with apocynin (ARM + APO) adjunctive based on scores of Rapid Murine Come behavior Scale (RMCBS). After a 30-day survivability period, we performed Barnes maze, T-maze, and novel object recognition cognitive tests to evaluate working and reference memory in all the experimental groups except CM. Sensorimotor tests were conducted in all the cohorts to assess motor coordination. We performed Golgi-Cox staining to illustrate cornu ammonis-1 (CA1) pyramidal neuronal morphology and study overall hippocampal neuronal density changes. Further, expression of NOX2, NeuN (neuronal marker) in hippocampal CA1 and dentate gyrus was determined using double immunofluorescence experiments in all the experimental groups. Mice administered with ARM monotherapy and APO adjunctive treatment exhibited similar survivability. The latter showed better locomotor and cognitive functions, reduced ROS levels, and hippocampal NOX2 immunoreactivity in ECM. Our results show a substantial increase in hippocampal NeuN immunoreactivity and dendritic arborization in ARM + APO cohorts compared to ARM-treated brain samples. Overall, our study suggests that overexpression of NOX2 could result in loss of hippocampal neuronal density and dendritic spines of CA1 neurons affecting the spatial working and reference memory during ECM. Notably, ARM + APO adjunctive therapy reversed the altered neuronal morphology and oxidative damage in hippocampal neurons restoring long-term cognitive functions after CM.


Subject(s)
Cognitive Dysfunction , Malaria, Cerebral , Animals , Cognitive Dysfunction/complications , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Hippocampus/metabolism , Malaria, Cerebral/complications , Malaria, Cerebral/drug therapy , Malaria, Cerebral/metabolism , Mice , NADPH Oxidases/metabolism , Neurons/metabolism
15.
FASEB J ; 35(12): e21997, 2021 12.
Article in English | MEDLINE | ID: mdl-34719814

ABSTRACT

The deadliest complication of infection by Plasmodium parasites, cerebral malaria, accounts for the majority of malarial fatalities. Although our understanding of the cellular and molecular mechanisms underlying the pathology remains incomplete, recent studies support the contribution of systemic and neuroinflammation as the cause of cerebral edema and blood-brain barrier (BBB) dysfunction. All Plasmodium species encode an orthologue of the innate cytokine, Macrophage Migration Inhibitory Factor (MIF), which functions in mammalian biology to regulate innate responses. Plasmodium MIF (PMIF) similarly signals through the host MIF receptor CD74, leading to an enhanced inflammatory response. We investigated the PMIF-CD74 interaction in the onset of experimental cerebral malaria (ECM) and liver stage Plasmodium development by using a combination of CD74 deficient (Cd74-/- ) hosts and PMIF deficient parasites. Cd74-/- mice were found to be protected from ECM and the protection was associated with the inability of brain microvessels to present parasite antigen to sequestered and pathogenic Plasmodium-specific CD8+ T cells. Infection of WT hosts with PMIF-deficient sporozoites or infection of Cd74-/- hosts with WT sporozoites impacted the survival of infected hepatocytes and subsequently reduced blood-stage associated inflammation, contributing to protection from ECM. We recapitulated these finding with a novel pharmacologic PMIF-selective antagonist that reduced PMIF/CD74 signaling and fully protected mice from ECM. These findings reveal a conserved mechanism for Plasmodium usurpation of host CD74 signaling and suggest a tractable approach for new pharmacologic intervention.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/chemistry , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class II/chemistry , Inflammation/prevention & control , Liver/pathology , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Malaria, Cerebral/prevention & control , Plasmodium berghei/physiology , Animals , Antigens, Differentiation, B-Lymphocyte/physiology , Histocompatibility Antigens Class II/physiology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Liver/immunology , Liver/parasitology , Macrophage Migration-Inhibitory Factors/metabolism , Malaria, Cerebral/etiology , Malaria, Cerebral/metabolism , Malaria, Cerebral/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
Front Immunol ; 12: 719189, 2021.
Article in English | MEDLINE | ID: mdl-34456927

ABSTRACT

Epidemiological studies provide compelling evidence that glucose-6-phosphate dehydrogenase (G6PD) deficiency individuals are relatively protected against Plasmodium parasite infection. However, the animal model studies on this subject are lacking. Plus, the underlying mechanism in vivo is poorly known. In this study, we used a G6pd-deficient mice infected with the rodent parasite Plasmodium berghei (P.berghei) to set up a malaria model in mice. We analyzed the pathological progression of experimental cerebral malaria (ECM) and acute liver injury in mice with different G6pd activity infected with P.berghei. We performed dual RNA-seq for host-parasite transcriptomics and validated the changes of proinflammatory response in the murine model. G6pd-deficient mice exhibited a survival advantage, less severe ECM and mild liver injury compared to the wild type mice. Analysis based on dual RNA-seq suggests that G6pd-deficient mice are protected from ECM and acute liver injury were related to proinflammatory responses. Th1 differentiation and dendritic cell maturation in the liver and spleen were inhibited in G6pd-deficient mice. The levels of proinflammatory cytokines were reduced, chemokines and vascular adhesion molecules in the brain were significantly down-regulated, these led to decreased cerebral microvascular obstruction in G6pd-deficient mice. We generated the result that G6pd-deficiency mediated protection against ECM and acute liver injury were driven by the regulatory proinflammatory responses. Furthermore, bioinformatics analyses showed that P.berghei might occur ribosome loss in G6pd-deficient mice. Our findings provide a novel perspective of the underlying mechanism of G6PD deficiency mediated protection against malaria in vivo.


Subject(s)
Glucosephosphate Dehydrogenase Deficiency/complications , Glucosephosphate Dehydrogenase Deficiency/metabolism , Glucosephosphate Dehydrogenase/metabolism , Liver Diseases, Parasitic/complications , Liver Diseases, Parasitic/prevention & control , Malaria, Cerebral/complications , Malaria, Cerebral/prevention & control , Animals , Biomarkers , Biopsy , Blood-Brain Barrier/metabolism , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Enzyme Activation , Gene Expression Profiling , Glucosephosphate Dehydrogenase Deficiency/etiology , Hemolysis , Inflammation Mediators/metabolism , Liver Diseases, Parasitic/metabolism , Liver Diseases, Parasitic/pathology , Malaria, Cerebral/metabolism , Mice , Plasmodium berghei
17.
Immunology ; 164(4): 737-753, 2021 12.
Article in English | MEDLINE | ID: mdl-34407221

ABSTRACT

Experimental cerebral malaria (ECM) is a severe complication of Plasmodium berghei ANKA (PbA) infection in mice, characterized by CD8+ T-cell accumulation within the brain. Whilst the dynamics of CD8+ T-cell activation and migration during extant primary PbA infection have been extensively researched, the fate of the parasite-specific CD8+ T cells upon resolution of ECM is not understood. In this study, we show that memory OT-I cells persist systemically within the spleen, lung and brain following recovery from ECM after primary PbA-OVA infection. Whereas memory OT-I cells within the spleen and lung exhibited canonical central memory (Tcm) and effector memory (Tem) phenotypes, respectively, memory OT-I cells within the brain post-PbA-OVA infection displayed an enriched CD69+ CD103- profile and expressed low levels of T-bet. OT-I cells within the brain were excluded from short-term intravascular antibody labelling but were targeted effectively by longer-term systemically administered antibodies. Thus, the memory OT-I cells were extravascular within the brain post-ECM but were potentially not resident memory cells. Importantly, whilst memory OT-I cells exhibited strong reactivation during secondary PbA-OVA infection, preventing activation of new primary effector T cells, they had dampened reactivation during a fourth PbA-OVA infection. Overall, our results demonstrate that memory CD8+ T cells are systemically distributed but exhibit a unique phenotype within the brain post-ECM, and that their reactivation characteristics are shaped by infection history. Our results raise important questions regarding the role of distinct memory CD8+ T-cell populations within the brain and other tissues during repeat Plasmodium infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Host-Parasite Interactions/immunology , Malaria/immunology , Malaria/parasitology , Plasmodium berghei/physiology , Animals , Biomarkers , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Chemotaxis, Leukocyte/immunology , Disease Susceptibility , Epitopes, T-Lymphocyte/immunology , Erythrocytes/immunology , Erythrocytes/parasitology , Extracellular Matrix , Immunologic Memory , Immunophenotyping , Life Cycle Stages , Lymphocyte Activation/immunology , Malaria/metabolism , Malaria/pathology , Malaria, Cerebral/immunology , Malaria, Cerebral/metabolism , Malaria, Cerebral/parasitology , Mice , Mice, Transgenic , Organ Specificity/immunology
18.
Int J Biol Macromol ; 177: 535-547, 2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33631268

ABSTRACT

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.


Subject(s)
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
19.
Acta Trop ; 217: 105867, 2021 May.
Article in English | MEDLINE | ID: mdl-33610534

ABSTRACT

Cerebral malaria is an outcome of multifaceted and complicated condition. Cytoadherence is one critical factor in cerebral malaria pathology as high order cytoadherence complexes result in vascular congestion and cell apoptosis. Morphological abnormalities in uninfected RBCs can be a contributing factor to aggravate cytoadherence. Malaria pigment hemozoin is a potential bioactive molecule and the role of this pigment in cerebral malaria pathology is not completely understood. To understand this, primarily we investigated the impact of hemozoin pigment on uninfected RBCs. Secondarily, we investigated the role of this pigment in formation of endothelial cells-RBCs (EC-RBC) cytoadherence complex. We first observed that a dose dependent hemozoin exposure to uninfected RBCs induced structural abnormalities. Differential counting of these abnormal RBCs indicated population of acanthocytes, spherocytes and microcytes. The formation of abnormal RBCs was observed with phosphatidylserine externalization. Lipid peroxidation, reduced glutathione and reactive oxygen species (ROS) levels indicated an increase in hemozoin exposure mediated oxidative stress. Our in-vitro cytoadherence assay indicated formation of endothelial EC-RBC cytoadherence complex. The dose dependent hemozoin exposure to uninfected RBCs resulted in oxidative stress mediated high order cytoadherence complex formation. This effect was reversed in presence of antioxidant molecules. The inhibitory effect of antioxidant molecules indicates that oxidative stress can be a regulatory factor to control cerebral malaria pathology. Being the first report to highlight the impact of malaria pigment hemozoin on uninfected RBCs, this study brings attention to the role of abnormal RBCs in worsening of cerebral malaria pathology.


Subject(s)
Endothelial Cells/pathology , Erythrocytes/pathology , Hemeproteins/metabolism , Malaria, Cerebral/metabolism , Malaria, Cerebral/pathology , Erythrocytes/parasitology , Humans , Lipid Peroxidation , Oxidative Stress , Plasmodium falciparum/metabolism , Plasmodium falciparum/physiology , Reactive Oxygen Species/metabolism
20.
Mediators Inflamm ; 2020: 3280689, 2020.
Article in English | MEDLINE | ID: mdl-32801995

ABSTRACT

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.


Subject(s)
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
SELECTION OF CITATIONS
SEARCH DETAIL
...