Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.431
Filtrar
1.
Trends Parasitol ; 40(1): 28-44, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38065791

RESUMO

Cerebral malaria (CM) is a severe neurological complication caused by Plasmodium falciparum parasites; it is characterized by the sequestration of infected red blood cells within the cerebral microvasculature. New findings, combined with a better understanding of the central nervous system (CNS) barriers, have provided greater insight into the players and events involved in CM, including site-specific T cell responses in the human brain. Here, we review the updated roles of innate and adaptive immune responses in CM, with a focus on the role of the perivascular macrophage-endothelium unit in antigen presentation, in the vascular and perivascular compartments. We suggest that these events may be pivotal in the development of CM.


Assuntos
Malária Cerebral , Humanos , Encéfalo , Plasmodium falciparum/fisiologia , Interações Hospedeiro-Parasita , Eritrócitos/parasitologia
2.
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
3.
PLoS Pathog ; 19(12): e1011807, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38051755

RESUMO

Malaria is caused by the rapid proliferation of Plasmodium parasites in patients and disease severity correlates with the number of infected red blood cells in circulation. Parasite multiplication within red blood cells is called schizogony and occurs through an atypical multinucleated cell division mode. The mechanisms regulating the number of daughter cells produced by a single progenitor are poorly understood. We investigated underlying regulatory principles by quantifying nuclear multiplication dynamics in Plasmodium falciparum and knowlesi using super-resolution time-lapse microscopy. This confirmed that the number of daughter cells was consistent with a model in which a counter mechanism regulates multiplication yet incompatible with a timer mechanism. P. falciparum cell volume at the start of nuclear division correlated with the final number of daughter cells. As schizogony progressed, the nucleocytoplasmic volume ratio, which has been found to be constant in all eukaryotes characterized so far, increased significantly, possibly to accommodate the exponentially multiplying nuclei. Depleting nutrients by dilution of culture medium caused parasites to produce fewer merozoites and reduced proliferation but did not affect cell volume or total nuclear volume at the end of schizogony. Our findings suggest that the counter mechanism implicated in malaria parasite proliferation integrates extracellular resource status to modify progeny number during blood stage infection.


Assuntos
Malária Falciparum , Malária , Parasitos , Animais , Humanos , Parasitos/fisiologia , Malária Falciparum/parasitologia , Malária/parasitologia , Plasmodium falciparum/fisiologia , Merozoítos/fisiologia , Eritrócitos/parasitologia
4.
PLoS Pathog ; 19(10): e1011661, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37856470

RESUMO

The pathophysiology of severe falciparum malaria involves a complex interaction between the host, parasite, and gut microbes. In this review, we focus on understanding parasite-induced intestinal injury and changes in the human intestinal microbiota composition in patients with Plasmodium falciparum malaria. During the blood stage of P. falciparum infection, infected red blood cells adhere to the vascular endothelium, leading to widespread microcirculatory obstruction in critical tissues, including the splanchnic vasculature. This process may cause intestinal injury and gut leakage. Epidemiological studies indicate higher rates of concurrent bacteraemia in severe malaria cases. Furthermore, severe malaria patients exhibit alterations in the composition and diversity of the intestinal microbiota, although the exact contribution to pathophysiology remains unclear. Mouse studies have demonstrated that the gut microbiota composition can impact susceptibility to Plasmodium infections. In patients with severe malaria, the microbiota shows an enrichment of pathobionts, including pathogens that are known to cause concomitant bloodstream infections. Microbial metabolites have also been detected in the plasma of severe malaria patients, potentially contributing to metabolic acidosis and other clinical complications. However, establishing causal relationships requires intervention studies targeting the gut microbiota.


Assuntos
Microbioma Gastrointestinal , Enteropatias , Malária Falciparum , Malária , Humanos , Animais , Camundongos , Microcirculação , Malária Falciparum/parasitologia , Malária/parasitologia , Plasmodium falciparum/fisiologia
5.
Elife ; 122023 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-37796723

RESUMO

Basigin is an essential host receptor for invasion of Plasmodium falciparum into human erythrocytes, interacting with parasite surface protein PfRH5. PfRH5 is a leading blood-stage malaria vaccine candidate and a target of growth-inhibitory antibodies. Here, we show that erythrocyte basigin is exclusively found in one of two macromolecular complexes, bound either to plasma membrane Ca2+-ATPase 1/4 (PMCA1/4) or to monocarboxylate transporter 1 (MCT1). PfRH5 binds to each of these complexes with a higher affinity than to isolated basigin ectodomain, making it likely that these are the physiological targets of PfRH5. PMCA-mediated Ca2+ export is not affected by PfRH5, making it unlikely that this is the mechanism underlying changes in calcium flux at the interface between an erythrocyte and the invading parasite. However, our studies rationalise the function of the most effective growth-inhibitory antibodies targeting PfRH5. While these antibodies do not reduce the binding of PfRH5 to monomeric basigin, they do reduce its binding to basigin-PMCA and basigin-MCT complexes. This indicates that the most effective PfRH5-targeting antibodies inhibit growth by sterically blocking the essential interaction of PfRH5 with basigin in its physiological context.


Assuntos
Malária Falciparum , Plasmodium falciparum , Humanos , Plasmodium falciparum/fisiologia , Basigina , Eritrócitos/parasitologia , Anticorpos Neutralizantes , Malária Falciparum/parasitologia , Proteínas de Protozoários/metabolismo , Ligação Proteica , Antígenos de Protozoários
6.
Science ; 381(6657): 533-540, 2023 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-37535741

RESUMO

Malaria control demands the development of a wide range of complementary strategies. We describe the properties of a naturally occurring, non-genetically modified symbiotic bacterium, Delftia tsuruhatensis TC1, which was isolated from mosquitoes incapable of sustaining the development of Plasmodium falciparum parasites. D. tsuruhatensis TC1 inhibits early stages of Plasmodium development and subsequent transmission by the Anopheles mosquito through secretion of a small-molecule inhibitor. We have identified this inhibitor to be the hydrophobic molecule harmane. We also found that, on mosquito contact, harmane penetrates the cuticle, inhibiting Plasmodium development. D. tsuruhatensis TC1 stably populates the mosquito gut, does not impose a fitness cost on the mosquito, and inhibits Plasmodium development for the mosquito's life. Contained field studies in Burkina Faso and modeling showed that D. tsuruhatensis TC1 has the potential to complement mosquito-targeted malaria transmission control.


Assuntos
Anopheles , Delftia , Interações Hospedeiro-Parasita , Malária Falciparum , Plasmodium falciparum , Animais , Anopheles/microbiologia , Malária Falciparum/microbiologia , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Plasmodium falciparum/microbiologia , Plasmodium falciparum/fisiologia , Delftia/fisiologia , Simbiose , Humanos
7.
J Biol Chem ; 299(9): 105111, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37517694

RESUMO

Upon infection by the malaria parasite Plasmodium falciparum, the glycolytic rate of a red blood cell increases up to 100-fold, possibly contributing to lactic acidosis and hypoglycemia in patients with severe malaria. This dramatic increase in glucose uptake and metabolism was correctly predicted by a newly constructed detailed enzyme kinetic model of glucose metabolism in the trophozoite-infected red blood cell. Subsequently, we expanded the model to simulate an infected red blood cell culture, including the different asexual blood-stage forms of the malaria parasite. The model simulations were in good agreement with experimental data, for which the measured parasitic volume was an important parameter. Upon further analysis of the model, we identified glucose transport as a drug target that would specifically affect infected red blood cells, which was confirmed experimentally with inhibitor titrations. This model can be a first step in constructing a whole-body model for glucose metabolism in malaria patients to evaluate the contribution of the parasite's metabolism to the disease state.


Assuntos
Antimaláricos , Eritrócitos , Glicólise , Malária Falciparum , Modelos Biológicos , Terapia de Alvo Molecular , Plasmodium falciparum , Humanos , Acidose Láctica , Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , Antimaláricos/metabolismo , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Glucose/metabolismo , Glicólise/efeitos dos fármacos , Hipoglicemia , Cinética , Malária Falciparum/metabolismo , Malária Falciparum/parasitologia , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidade , Plasmodium falciparum/fisiologia , Trofozoítos/patogenicidade , Trofozoítos/fisiologia , Terapia de Alvo Molecular/métodos , Carga Parasitária
8.
PLoS Pathog ; 19(6): e1011448, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37339122

RESUMO

Insecticide resistance is under strong selective pressure in Anopheles mosquitoes due to widespread usage of insecticides in vector control strategies. Resistance mechanisms likely cause changes that profoundly affect mosquito physiology, yet it remains poorly understood how selective pressures imposed by insecticides may alter the ability of the mosquito to host and transmit a Plasmodium infection. From pyrethroid-resistant field-derived Anopheles gambiae s.l. mosquitoes, we established resistant (RES) and susceptible (SUS) colonies by either selection for, or loss of insecticide resistance. We show increased oocyst intensity and growth rate as well as increased sporozoite prevalence and intensity in RES compared to SUS females infected with Plasmodium falciparum. The increase in infection intensity in RES females was not associated with the presence of the kdrL1014F mutation and was not impacted by inhibition of Cytochrome P450s. The lipid transporter lipophorin (Lp), which was upregulated in RES compared to SUS, was at least partly implicated in the increased intensity of P. falciparum but not directly involved in the insecticide resistance phenotype. Interestingly, we observed that although P. falciparum infections were not affected when RES females were exposed to permethrin, these females had decreased lipid abundance in the fat body following exposure, pointing to a possible role for lipid mobilization in response to damage caused by insecticide challenge. The finding that selection for insecticide resistance can increase P. falciparum infection intensities and growth rate reinforces the need to assess the overall impact on malaria transmission dynamics caused by selective pressures mosquitoes experience during repeated insecticide challenge.


Assuntos
Anopheles , Inseticidas , Malária Falciparum , Malária , Animais , Feminino , Inseticidas/farmacologia , Plasmodium falciparum/fisiologia , Resistência a Inseticidas/genética , Anopheles/fisiologia , Mosquitos Vetores/genética , Lipídeos , Controle de Mosquitos
9.
Elife ; 122023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36916164

RESUMO

The malaria parasite life cycle includes asexual replication in human blood, with a proportion of parasites differentiating to gametocytes required for transmission to mosquitoes. Commitment to differentiate into gametocytes, which is marked by activation of the parasite transcription factor ap2-g, is known to be influenced by host factors but a comprehensive model remains uncertain. Here, we analyze data from 828 children in Kilifi, Kenya with severe, uncomplicated, and asymptomatic malaria infection over 18 years of falling malaria transmission. We examine markers of host immunity and metabolism, and markers of parasite growth and transmission investment. We find that inflammatory responses associated with reduced plasma lysophosphatidylcholine levels are associated with markers of increased investment in parasite sexual reproduction (i.e. transmission investment) and reduced growth (i.e. asexual replication). This association becomes stronger with falling transmission and suggests that parasites can rapidly respond to the within-host environment, which in turn is subject to changing transmission.


Assuntos
Malária Falciparum , Malária , Parasitos , Animais , Criança , Humanos , Plasmodium falciparum/fisiologia , Malária/parasitologia , Reprodução , Adaptação Fisiológica , Malária Falciparum/parasitologia
10.
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
11.
Microbes Infect ; 25(5): 105102, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36708871

RESUMO

To persist in the blood circulation and to be available for mosquitoes, Plasmodium falciparum gametocytes modify the deformability and the permeability of their erythrocyte host via cyclic AMP (cAMP) signaling pathway. Cyclic nucleotide levels are tightly controlled by phosphodiesterases (PDE), however in Plasmodium these proteins are poorly characterized. Here, we characterize the P. falciparum phosphodiesterase delta (PfPDEδ) and we investigate its role in the cAMP signaling-mediated regulation of gametocyte-infected erythrocyte mechanical properties. Our results revealed that PfPDEδ is a dual-function enzyme capable of hydrolyzing both cAMP and cGMP, with a higher affinity for cAMP. We also show that PfPDEδ is the most expressed PDE in mature gametocytes and we propose that it is located in parasitophorous vacuole at the interface between the host cell and the parasite. We conclude that PfPDEδ is the master regulator of both the increase in deformability and the inhibition of channel activity in mature gametocyte stages, and may therefore play a crucial role in the persistence of mature gametocytes in the bloodstream.


Assuntos
Malária Falciparum , Plasmodium falciparum , Animais , Plasmodium falciparum/fisiologia , Diester Fosfórico Hidrolases , Malária Falciparum/parasitologia , Eritrócitos/parasitologia , Transdução de Sinais
12.
Parasit Vectors ; 15(1): 434, 2022 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-36397133

RESUMO

BACKGROUND: Babesia is an intraerythrocytic parasite often misdiagnosed as a malaria parasite, leading to inappropriate treatment of the disease especially in co-endemic areas. In recent years, optical diffraction tomography (ODT) has shown great potential in the field of pathogen detection by quantification of three-dimensional (3D) imaging tomograms. The 3D imaging of biological cells is crucial to investigate and provide valuable information about the mechanisms behind the pathophysiology of cells and tissues. METHODS: The early ring stage of P. falciparum were obtained from stored stock of infected RBCs and of B. microti were obtained from infected patients during diagnosis. The ODT technique was applied to analyze and characterize detailed differences between P. falciparum and B. microti ring stage at the single cell level. Based on 3D quantitative information, accurate measurement was performed of morphological, biochemical, and biophysical parameters. RESULTS: Accurate measurements of morphological parameters indicated that the host cell surface area at the ring stage in B. microti was significantly smaller (140.2 ± 17.1 µm2) than that in P. falciparum (159.0 ± 15.2 µm2), and sphericities showed higher levels in B. microti-parasitized cells (0.66 ± 0.05) than in P. falciparum (0.60 ± 0.04). Based on biochemical parameters, host cell hemoglobin level was significantly higher and membrane fluctuations were respectively more active in P. falciparum-infected cells (30.25 ± 2.96 pg; 141.3 ± 24.68 nm) than in B. microti (27.28 ± 3.52 pg; 110.1 ± 38.83 nm). The result indicates that P. falciparum more actively altered host RBCs than B. microti. CONCLUSION: Although P. falciparum and B. microti often show confusable characteristics under the microscope, and the actual three-dimensional properties are different. These differences could be used in differential clinical diagnosis of erythrocytes infected with B. microti and P. falciparum.


Assuntos
Babesia microti , Babesia , Malária Falciparum , Humanos , Plasmodium falciparum/fisiologia , Eritrócitos/parasitologia
13.
Adv Sci (Weinh) ; 9(36): e2202944, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36300890

RESUMO

Reorganization of host red blood cells by the malaria parasite Plasmodium falciparum enables their sequestration via attachment to the microvasculature. This artificially increases the dwelling time of the infected red blood cells within inner organs such as the brain, which can lead to cerebral malaria. Cerebral malaria is the deadliest complication patients infected with P. falciparum can experience and still remains a major public health concern despite effective antimalarial therapies. Here, the current understanding of the effect of P. falciparum cytoadherence and their secreted proteins on structural features of the human blood-brain barrier and their involvement in the pathogenesis of cerebral malaria are highlighted. Advanced 2D and 3D in vitro models are further assessed to study this devastating interaction between parasite and host. A better understanding of the molecular mechanisms leading to neuronal and cognitive deficits in cerebral malaria will be pivotal in devising new strategies to treat and prevent blood-brain barrier dysfunction and subsequent neurological damage in patients with cerebral malaria.


Assuntos
Malária Cerebral , Malária Falciparum , Humanos , Malária Cerebral/parasitologia , Malária Cerebral/patologia , Plasmodium falciparum/fisiologia , Malária Falciparum/metabolismo , Malária Falciparum/parasitologia , Encéfalo/patologia , Eritrócitos/metabolismo
14.
mBio ; 13(5): e0093722, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-35972144

RESUMO

The repeated emergence of antimalarial drug resistance in Plasmodium falciparum, including to the current frontline antimalarial artemisinin, is a perennial problem for malaria control. Next-generation sequencing has greatly accelerated the identification of polymorphisms in resistance-associated genes but has also highlighted the need for more sensitive and accurate laboratory tools to profile current and future antimalarials and to quantify the impact of drug resistance acquisition on parasite fitness. The interplay of fitness and drug response is of fundamental importance in understanding why particular genetic backgrounds are better at driving the evolution of drug resistance in natural populations, but the impact of parasite fitness landscapes on the epidemiology of drug resistance has typically been laborious to accurately quantify in the lab, with assays being limited in accuracy and throughput. Here we present a scalable method to profile fitness and drug response of genetically distinct P. falciparum strains with well-described sensitivities to several antimalarials. We leverage CRISPR/Cas9 genome-editing and barcode sequencing to track unique barcodes integrated into a nonessential gene (pfrh3). We validate this approach in multiplex competitive growth assays of three strains with distinct geographical origins. Furthermore, we demonstrate that this method can be a powerful approach for tracking artemisinin response as it can identify an artemisinin resistant strain within a mix of multiple parasite lines, suggesting an approach for scaling the laborious ring-stage survival assay across libraries of barcoded parasite lines. Overall, we present a novel high-throughput method for multiplexed competitive growth assays to evaluate parasite fitness and drug response. IMPORTANCE The complex interplay between antimalarial resistance and parasite fitness has important implications for understanding the development and spread of drug resistance alleles and the impact of genetic background on transmission. One limitation with current methodologies to measure parasite fitness is the ability to scale this beyond simple head-to-head competition experiments between a wildtype control line and test line, with a need for a scalable approach that allows tracking of parasite growth in complex mixtures. In our study, we have used CRISPR editing to insert unique DNA barcodes into a safe-harbor genomic locus to tag multiple parasite strains and use next-generation sequencing to read out strain dynamics. We observe inherent fitness differences between the strains, as well as sensitive modulation of responses to challenge with clinically relevant antimalarials, including artemisinin.


Assuntos
Antimaláricos , Artemisininas , Plasmodium falciparum , Antimaláricos/farmacologia , Artemisininas/farmacologia , Misturas Complexas , Resistência a Medicamentos/genética , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/genética , Aptidão Genética
15.
Methods Mol Biol ; 2470: 493-503, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35881369

RESUMO

Rosetting is the ability of Plasmodium falciparum-infected erythrocytes (IEs) to bind to host receptors on the surface of uninfected erythrocytes (uE) leading to the formation of a cluster of cells with a central IE surrounded by uE. It is a hallmark event during the pathogenesis of P. falciparum malaria, the most severe species causing malaria, which affects mostly young children in Africa. There are no current treatments effectively targeting and disrupting parasite rosette formation. Here, we detail a high-throughput, flow cytometry based assay that allows testing and identification of potential rosetting-inhibitory compounds that could be used in combination with anti-plasmodial drugs to reduce malaria morbidity and mortality.


Assuntos
Malária Falciparum , Parasitos , Animais , Criança , Pré-Escolar , Eritrócitos/metabolismo , Citometria de Fluxo , Humanos , Malária Falciparum/parasitologia , Plasmodium falciparum/fisiologia , Formação de Roseta
16.
Methods Mol Biol ; 2470: 545-556, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35881374

RESUMO

Unique to Plasmodium falciparum malaria parasites, the mature asexual stages of the life cycle are absent from the peripheral blood stream. Using syringe pumps and commercially available microslides, it is possible to mimic the blood flow, and investigate the interactions of erythrocytes infected by well-defined P. falciparum isolates for their ability to bind to various tissue receptors under physiological flow conditions. This chapter outlines the techniques needed to investigate how parasites bind to endothelial cells under physiological sheer stress conditions.


Assuntos
Malária Falciparum , Plasmodium falciparum , Adesão Celular/fisiologia , Células Endoteliais , Eritrócitos/parasitologia , Humanos , Malária Falciparum/parasitologia , Plasmodium falciparum/fisiologia
17.
Elife ; 112022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35775739

RESUMO

Malaria is caused by infection of the erythrocytes by the parasites Plasmodium. Inside the erythrocytes, the parasites multiply via schizogony, an unconventional cell division mode. The inner membrane complex (IMC), an organelle located beneath the parasite plasma membrane, serving as the platform for protein anchorage, is essential for schizogony. So far, the complete repertoire of IMC proteins and their localization determinants remain unclear. Here we used biotin ligase (TurboID)-based proximity labeling to compile the proteome of the schizont IMC of the rodent malaria parasite Plasmodium yoelii. In total, 300 TurboID-interacting proteins were identified. 18 of 21 selected candidates were confirmed to localize in the IMC, indicating good reliability. In light of the existing palmitome of Plasmodium falciparum, 83 proteins of the P. yoelii IMC proteome are potentially palmitoylated. We further identified DHHC2 as the major resident palmitoyl-acyl-transferase of the IMC. Depletion of DHHC2 led to defective schizont segmentation and growth arrest both in vitro and in vivo. DHHC2 was found to palmitoylate two critical IMC proteins CDPK1 and GAP45 for their IMC localization. In summary, this study reports an inventory of new IMC proteins and demonstrates a central role of DHHC2 in governing the IMC localization of proteins during the schizont development.


Assuntos
Malária , Parasitos , Animais , Eritrócitos/parasitologia , Lipoilação , Malária/parasitologia , Parasitos/metabolismo , Plasmodium falciparum/fisiologia , Proteoma/metabolismo , Proteômica , Proteínas de Protozoários/metabolismo , Reprodutibilidade dos Testes , Esquizontes
18.
Mol Biol Rep ; 49(11): 10579-10591, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35670928

RESUMO

BACKGROUND: Cerebral malaria is often pronounced as a major life-threatening neurological complication of Plasmodium falciparum infection. The complex pathogenic landscape of the parasite and the associated neurological complications are still not elucidated properly. The growing concerns of drugresistant parasite strains along with the failure of anti-malarial drugs to subdue post-recovery neuro-cognitive dysfunctions in cerebral malaria patients have called for a demand to explore novel biomarkers and therapeutic avenues. Due course of the brain infection journey of the parasite, events such as sequestration of infected RBCs, cytoadherence, inflammation, endothelial activation, and blood-brain barrier disruption are considered critical. METHODS: In this review, we briefly summarize the diverse pathogenesis of the brain-invading parasite associated with loss of the blood-brain barrier integrity. In addition, we also discuss proteomics, transcriptomics, and bioinformatics strategies to identify an array of new biomarkers and drug candidates. CONCLUSION: A proper understanding of the parasite biology and mechanism of barrier disruption coupled with emerging state-of-art therapeutic approaches could be helpful to tackle cerebral malaria.


Assuntos
Malária Cerebral , Malária Falciparum , Humanos , Malária Cerebral/tratamento farmacológico , Malária Cerebral/parasitologia , Malária Cerebral/patologia , Plasmodium falciparum/fisiologia , Barreira Hematoencefálica/patologia , Biomarcadores
19.
Curr Neurol Neurosci Rep ; 22(8): 499-513, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35699901

RESUMO

PURPOSE OF REVIEW: To discuss the neurological complications and pathophysiology of organ damage following malaria infection. RECENT FINDINGS: The principal advancement made in malaria research has been a better understanding of the pathogenesis of cerebral malaria (CM), the most dreaded neurological complication generally caused by Plasmodium falciparum infection. However, no definitive treatment has yet been evolved other than the use of antimalarial drugs and supportive care. The development of severe cerebral edema in CM results from two distinct pathophysiologic mechanisms. First, the development of "sticky" red blood cells (RBCs) leads to cytoadherence, where red blood cells (RBCs) get stuck to the endothelial walls and between themselves, resulting in clogging of the brain microvasculature with resultant hypoxemia and cerebral edema. In addition, the P. falciparum-infected erythrocyte membrane protein 1 (PfEMP1) molecules protrude from the raised knob structures on the RBCs walls and are in themselves made of a combination of human and parasite proteins in a tight complex. Antibodies to surfins, rifins, and stevors from the parasite are also located in the RBC membrane. On the human microvascular side, a range of molecules involved in host-parasite interactions, including CD36 and intracellular adhesion molecule 1, is activated during interaction with other molecules such as endothelial protein C receptor and thrombospondin. As a result, an inflammatory response occurs with the dysregulated release of cytokines (TNF, interleukins 1 and 10) which damage the blood-brain barrier (BBB), causing plasma leakage and brain edema. This second mechanism of CNS injury often involves multiple organs in adult patients in endemic areas but remains localized only to the central nervous system (CNS) among African children. Neurological sequelae may follow both P. falciparum and P. vivax infections. The major brain pathology of CM is brain edema with diffuse brain swelling resulting from the combined effects of reduced perfusion and hypoxemia of cerebral neurons due to blockage of the microvasculature by parasitized RBCs as well as the neurotoxic effect of released cytokines from a hyper-acute immune host reaction. A plethora of additional neurological manifestations have been associated with malaria, including posterior reversible encephalopathy syndrome (PRES), reversible cerebral vasoconstriction syndrome (RCVS), malarial retinopathy, post-malarial neurological syndrome (PMNS), acute disseminated encephalomyelitis (ADEM), Guillain-Barré syndrome (GBS), and cerebellar ataxia. Lastly, the impact of the COVID-19 pandemic on worldwide malaria control programs and the possible threat from co-infections is briefly discussed.


Assuntos
Edema Encefálico , COVID-19 , Malária Cerebral , Malária Falciparum , Síndrome da Leucoencefalopatia Posterior , Adulto , Criança , Citocinas , Humanos , Hipóxia , Malária Cerebral/complicações , Malária Cerebral/parasitologia , Malária Falciparum/complicações , Malária Falciparum/parasitologia , Pandemias , Plasmodium falciparum/fisiologia
20.
J Proteome Res ; 21(8): 1857-1867, 2022 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-35772009

RESUMO

Histones are the building units of nucleosomes, which constitute chromatin. Histone post-translational modifications (PTMs) play an essential role in epigenetic gene regulation. The Plasmodium falciparum genome encodes canonical and variant histones and a collection of conserved enzymes for histone PTMs and chromatin remodeling. Herein, we profiled the P. falciparum histone PTMs during the development of gametocytes, the obligatory stage for parasite transmission. Mass spectrometric analysis of histones extracted from the early, middle, and late stages of gametocytes identified 457 unique histone peptides with 90 PTMs, of which 50% were novel. The gametocyte histone PTMs display distinct patterns from asexual stages, with many new methylation sites in histones H3 and H3.3 (e.g., K14, K18, and K37). Quantitative analyses revealed a high abundance of acetylation in H3 and H4, mono-methylation of H3/H3.3 K37, and ubiquitination of H3BK112, suggesting that these PTMs play critical roles in gametocytes. Gametocyte histones also showed extensive and unique combinations of PTMs. These data indicate that the parasite harbors distinct transcription regulation mechanisms during gametocyte development and lay the foundation for further characterization of epigenetic regulation in the life cycle of the malaria parasite.


Assuntos
Gametogênese , Histonas , Plasmodium falciparum , Processamento de Proteína Pós-Traducional , Acetilação , Epigênese Genética/genética , Gametogênese/genética , Gametogênese/fisiologia , Histonas/genética , Histonas/metabolismo , Humanos , Estágios do Ciclo de Vida/genética , Estágios do Ciclo de Vida/fisiologia , Malária Falciparum/parasitologia , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Plasmodium falciparum/fisiologia , Processamento de Proteína Pós-Traducional/genética , Processamento de Proteína Pós-Traducional/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...