Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 228
Filtrar
1.
Brain Res ; 1822: 148669, 2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37951562

RESUMO

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.


Assuntos
Barreira Hematoencefálica , Bradicinina , Adesão Celular , Malária Cerebral , Malária Falciparum , Plasmodium falciparum , Humanos , Bradicinina/metabolismo , Adesão Celular/fisiologia , Meios de Cultivo Condicionados/farmacologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Eritrócitos/parasitologia , Malária Cerebral/metabolismo , Malária Cerebral/parasitologia , Malária Falciparum/metabolismo , Malária Falciparum/parasitologia , Monócitos/fisiologia , Plasmodium falciparum/fisiologia , Barreira Hematoencefálica/fisiopatologia
2.
Mem Inst Oswaldo Cruz ; 118: e230033, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37403869

RESUMO

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.


Assuntos
Malária Cerebral , Malária Falciparum , Humanos , Plasmodium falciparum , Molécula 1 de Adesão Intercelular/metabolismo , Células Endoteliais , Molécula 1 de Adesão de Célula Vascular/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Malária Cerebral/metabolismo , Malária Cerebral/patologia , Monoterpenos/metabolismo , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Endotélio Vascular , Permeabilidade
3.
Cells ; 12(7)2023 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-37048057

RESUMO

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.


Assuntos
Malária Cerebral , Humanos , Malária Cerebral/metabolismo , Malária Cerebral/parasitologia , Malária Cerebral/patologia , Células Endoteliais/metabolismo , Reprodutibilidade dos Testes , Encéfalo/patologia , Plasmodium falciparum , Organoides/metabolismo
4.
Parasitol Res ; 122(3): 729-737, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36694092

RESUMO

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.


Assuntos
Células Endoteliais , Malária Cerebral , Humanos , Animais , Camundongos , Encéfalo/metabolismo , Barreira Hematoencefálica , Malária Cerebral/tratamento farmacológico , Malária Cerebral/metabolismo , Plasmodium falciparum/fisiologia
5.
PLoS One ; 18(1): e0280282, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36626386

RESUMO

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.


Assuntos
Malária Cerebral , Receptores de Complemento 3b , Humanos , Eritrócitos , Malária Cerebral/genética , Malária Cerebral/metabolismo , Papua Nova Guiné , Receptores de Complemento 3b/genética , Seleção Genética , Genética Populacional , Índia
6.
Mol Omics ; 18(8): 716-730, 2022 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-35960011

RESUMO

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.


Assuntos
Antimaláricos , Artemisininas , Malária Cerebral , Animais , Anti-Inflamatórios/uso terapêutico , Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , Artemisininas/farmacologia , Artemisininas/uso terapêutico , Artesunato/farmacologia , Artesunato/uso terapêutico , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Molécula 1 de Adesão Intercelular/uso terapêutico , Interleucina-10/uso terapêutico , Interleucina-6/uso terapêutico , Chumbo/uso terapêutico , Malária Cerebral/tratamento farmacológico , Malária Cerebral/genética , Malária Cerebral/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , RNA-Seq , Receptores Toll-Like/uso terapêutico , Fator de Necrose Tumoral alfa/uso terapêutico
7.
Mol Brain ; 15(1): 57, 2022 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-35725567

RESUMO

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.


Assuntos
Ferroptose , Malária Cerebral , Animais , Linfócitos T CD8-Positivos , Malária Cerebral/metabolismo , Malária Cerebral/patologia , Camundongos , Neurônios/metabolismo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase
8.
J Infect Dis ; 226(8): 1470-1479, 2022 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-35556124

RESUMO

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.


Assuntos
Malária Cerebral , Malária Falciparum , Encéfalo/patologia , Capilares/patologia , Criança , Epitopos/metabolismo , Eritrócitos/metabolismo , Glucosamina/metabolismo , Glicocálix/metabolismo , Glicosaminoglicanos/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Lectinas , Malária Cerebral/metabolismo , Manose/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Plasmodium falciparum/fisiologia
9.
EMBO Rep ; 23(6): e53641, 2022 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-35417070

RESUMO

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.


Assuntos
Malária Cerebral , Malária Falciparum , Plasmodium falciparum , Eritrócitos/parasitologia , Humanos , Malária Cerebral/genética , Malária Cerebral/metabolismo , Malária Falciparum/genética , Neutrófilos/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo
10.
Sci Adv ; 8(14): eabj7110, 2022 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-35385300

RESUMO

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.


Assuntos
Hipotireoidismo , Malária Cerebral , Sirtuína 1 , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Hipotireoidismo/metabolismo , Malária Cerebral/tratamento farmacológico , Malária Cerebral/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium berghei , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/metabolismo
11.
J Infect Dis ; 225(12): 2187-2196, 2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35255125

RESUMO

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.


Assuntos
Malária Cerebral , Malária Falciparum , Benin , Criança , Pré-Escolar , Eritrócitos/parasitologia , Perfilação da Expressão Gênica , Humanos , Malária Cerebral/metabolismo , Malária Falciparum/metabolismo , Plasmodium falciparum , Proteínas de Protozoários/metabolismo , Transcriptoma
12.
Mol Neurobiol ; 59(2): 800-820, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34782951

RESUMO

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.


Assuntos
Disfunção Cognitiva , Malária Cerebral , Animais , Disfunção Cognitiva/complicações , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Hipocampo/metabolismo , Malária Cerebral/complicações , Malária Cerebral/tratamento farmacológico , Malária Cerebral/metabolismo , Camundongos , NADPH Oxidases/metabolismo , Neurônios/metabolismo
13.
FASEB J ; 35(12): e21997, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34719814

RESUMO

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.


Assuntos
Antígenos de Diferenciação de Linfócitos B/química , Linfócitos T CD8-Positivos/imunologia , Antígenos de Histocompatibilidade Classe II/química , Inflamação/prevenção & controle , Fígado/patologia , Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Malária Cerebral/prevenção & controle , Plasmodium berghei/fisiologia , Animais , Antígenos de Diferenciação de Linfócitos B/fisiologia , Antígenos de Histocompatibilidade Classe II/fisiologia , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Fígado/imunologia , Fígado/parasitologia , Fatores Inibidores da Migração de Macrófagos/metabolismo , Malária Cerebral/etiologia , Malária Cerebral/metabolismo , Malária Cerebral/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
14.
Front Immunol ; 12: 719189, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34456927

RESUMO

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.


Assuntos
Deficiência de Glucosefosfato Desidrogenase/complicações , Deficiência de Glucosefosfato Desidrogenase/metabolismo , Glucosefosfato Desidrogenase/metabolismo , Hepatopatias Parasitárias/complicações , Hepatopatias Parasitárias/prevenção & controle , Malária Cerebral/complicações , Malária Cerebral/prevenção & controle , Animais , Biomarcadores , Biópsia , Barreira Hematoencefálica/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Ativação Enzimática , Perfilação da Expressão Gênica , Deficiência de Glucosefosfato Desidrogenase/etiologia , Hemólise , Mediadores da Inflamação/metabolismo , Hepatopatias Parasitárias/metabolismo , Hepatopatias Parasitárias/patologia , Malária Cerebral/metabolismo , Camundongos , Plasmodium berghei
15.
Immunology ; 164(4): 737-753, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34407221

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Interações Hospedeiro-Parasita/imunologia , Malária/imunologia , Malária/parasitologia , Plasmodium berghei/fisiologia , Animais , Biomarcadores , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Quimiotaxia de Leucócito/imunologia , Suscetibilidade a Doenças , Epitopos de Linfócito T/imunologia , Eritrócitos/imunologia , Eritrócitos/parasitologia , Matriz Extracelular , Memória Imunológica , Imunofenotipagem , Estágios do Ciclo de Vida , Ativação Linfocitária/imunologia , Malária/metabolismo , Malária/patologia , Malária Cerebral/imunologia , Malária Cerebral/metabolismo , Malária Cerebral/parasitologia , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos/imunologia
16.
Int J Biol Macromol ; 177: 535-547, 2021 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-33631268

RESUMO

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.


Assuntos
Malária Cerebral/genética , Malária Falciparum/genética , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Fatores de Virulência/genética , Humanos , Malária Cerebral/metabolismo , Malária Falciparum/metabolismo , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/metabolismo , Fatores de Virulência/metabolismo
17.
Acta Trop ; 217: 105867, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33610534

RESUMO

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.


Assuntos
Células Endoteliais/patologia , Eritrócitos/patologia , Hemeproteínas/metabolismo , Malária Cerebral/metabolismo , Malária Cerebral/patologia , Eritrócitos/parasitologia , Humanos , Peroxidação de Lipídeos , Estresse Oxidativo , Plasmodium falciparum/metabolismo , Plasmodium falciparum/fisiologia , Espécies Reativas de Oxigênio/metabolismo
18.
Mol Neurobiol ; 57(12): 5069-5083, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32833186

RESUMO

One-fourth survivors of cerebral malaria (CM) retain long-term cognitive and behavioral deficits. Structural abnormalities in striatum are reported in 80% of children with CM. Dopamine receptors (D1 and D2) are widely expressed in striatal medium spiny neurons (MSNs) that regulate critical physiological functions related to behavior and cognition. Dysregulation of dopamine receptors alters the expression of downstream proteins such as dopamine- and cAMP-regulated phosphoprotein (DARPP), Ca2+/calmodulin-dependent protein kinase II alpha (CaMKIIα), and p25/cyclin-dependent kinase 5 (cdk5). However, the role of dopamine receptor signaling dysfunction on the outcome of striatal neuron degeneration is unknown underlying the pathophysiology of CM. Using experimental CM (ECM), the present study attempted to understand the role of aberrant dopamine receptor signaling and its possible relation in causing MSNs morphological impairment. The effect of antimalarial drug artemether (ARM) rescue therapy was also assessed after ECM on the outcome of dopamine receptors downstream signaling. ECM was induced in C57BL/6 mice (male and female) infecting with Plasmodium berghei ANKA (PbA) parasite that reiterates the clinical setting of CM. We demonstrated that ECM caused a significant increase in the expression of D1, D2 receptors, phosphorylated DARPP, p25, cdk5, CaMKIIα, and D1-D2 heteromers. A substantial increase in neuronal damage observed in the dorsolateral striatum region of ECM brains (particularly in MSNs) as revealed by increased Fluoro-Jade C staining, reduced dendritic spine density, and impaired dendritic arborization with varicosities. While the ARM rescue therapy significantly altered the effects of ECM induced dopamine receptor signaling dysfunction and neurodegeneration. Overall, our data suggest that dysregulation of dopamine receptor signaling plays an important role in the degeneration of MSNs, and the ARM rescue therapy might provide better insights to develop effective therapeutic strategies for CM.


Assuntos
Corpo Estriado/patologia , Neurônios Dopaminérgicos/metabolismo , Malária Cerebral/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Transdução de Sinais , Animais , Comportamento Animal , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Feminino , Malária Cerebral/parasitologia , Masculino , Camundongos Endogâmicos C57BL , Parasitemia/patologia , Plasmodium berghei/fisiologia , Multimerização Proteica , Análise de Sobrevida
19.
Mediators Inflamm ; 2020: 3280689, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32801995

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Eritrócitos/metabolismo , Inflamação/sangue , Malária Cerebral/genética , Malária Cerebral/metabolismo , Transcriptoma/genética , Quimiocina CXCL10/sangue , Biologia Computacional/métodos , Humanos , Interleucina-18/sangue , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Proc Natl Acad Sci U S A ; 117(32): 19465-19474, 2020 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-32709745

RESUMO

Infection by malaria parasites triggers dynamic immune responses leading to diverse symptoms and pathologies; however, the molecular mechanisms responsible for these reactions are largely unknown. We performed Trans-species Expression Quantitative Trait Locus analysis to identify a large number of host genes that respond to malaria parasite infections. Here we functionally characterize one of the host genes called receptor transporter protein 4 (RTP4) in responses to malaria parasite and virus infections. RTP4 is induced by type I IFN (IFN-I) and binds to the TANK-binding kinase (TBK1) complex where it negatively regulates TBK1 signaling by interfering with expression and phosphorylation of both TBK1 and IFN regulatory factor 3. Rtp4-/- mice were generated and infected with malaria parasite Plasmodiun berghei ANKA. Significantly higher levels of IFN-I response in microglia, lower parasitemia, fewer neurologic symptoms, and better survival rates were observed in Rtp4-/- than in wild-type mice. Similarly, RTP4 deficiency significantly reduced West Nile virus titers in the brain, but not in the heart and the spleen, of infected mice, suggesting a specific role for RTP4 in brain infection and pathology. This study reveals functions of RTP4 in IFN-I response and a potential target for therapy in diseases with neuropathology.


Assuntos
Encéfalo/patologia , Interferon Tipo I/metabolismo , Malária Cerebral/patologia , Chaperonas Moleculares/metabolismo , Animais , Encéfalo/parasitologia , Encéfalo/virologia , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Fator Regulador 3 de Interferon , Malária Cerebral/metabolismo , Malária Cerebral/parasitologia , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Chaperonas Moleculares/genética , Fosforilação , Plasmodium berghei/fisiologia , Plasmodium yoelii/fisiologia , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Febre do Nilo Ocidental/metabolismo , Febre do Nilo Ocidental/patologia , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...