Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.433
Filtrar
1.
Malar J ; 23(1): 130, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38693572

RESUMEN

BACKGROUND: The sequestration of Plasmodium falciparum infected erythrocytes in the placenta, and the resulting inflammatory response affects maternal and child health. Despite existing information, little is known about the direct impact of P. falciparum on the placental barrier formed by trophoblast and villous stroma. This study aimed to assess placental tissue damage caused by P. falciparum in human placental explants (HPEs). METHODS: HPEs from chorionic villi obtained of human term placentas (n = 9) from normal pregnancies were exposed to P. falciparum-infected erythrocytes (IE) for 24 h. HPEs were embedded in paraffin blocks and used to study tissue damage through histopathological and histochemical analysis and apoptosis using TUNEL staining. Culture supernatants were collected to measure cytokine and angiogenic factors and to determine LDH activity as a marker of cytotoxicity. A subset of archived human term placenta paraffin-embedded blocks from pregnant women with malaria were used to confirm ex vivo findings. RESULTS: Plasmodium falciparum-IE significantly damages the trophoblast layer and the villous stroma of the chorionic villi. The increased LDH activity and pathological findings such as syncytial knots, fibrin deposits, infarction, trophoblast detachment, and collagen disorganization supported these findings. The specific damage to the trophoblast and the thickening of the subjacent basal lamina were more pronounced in the ex vivo infection. In contrast, apoptosis was higher in the in vivo infection. This disparity could be attributed to the duration of exposure to the infection, which significantly varied between individuals naturally exposed over time and the 24-h exposure in the ex vivo HPE model. CONCLUSION: Exposure to P. falciparum-IE induces a detachment of the syncytiotrophoblast, disorganization of the stroma villi, and an increase in apoptosis, alterations that may be associated with adverse results such as intrauterine growth restriction and low birth weight.


Asunto(s)
Vellosidades Coriónicas , Plasmodium falciparum , Trofoblastos , Humanos , Femenino , Vellosidades Coriónicas/parasitología , Vellosidades Coriónicas/patología , Embarazo , Plasmodium falciparum/fisiología , Trofoblastos/parasitología , Apoptosis , Malaria Falciparum/parasitología , Malaria Falciparum/patología , Placenta/parasitología , Placenta/patología , Citocinas/metabolismo
2.
Malar J ; 23(1): 135, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38711028

RESUMEN

BACKGROUND: The direct membrane feeding assay (DMFA), whereby gametocyte-infected blood is collected from human donors and from which mosquitoes feed through a membrane, is proving essential for assessing parameters influencing Plasmodium transmission potential in endemic countries. The success of DMFAs is closely tied to gametocyte density in the blood, with relatively high gametocytaemia ensuring optimal infection levels in mosquitoes. As transmission intensity declines with control efforts, the occurrence of asymptomatic individuals with low gametocyte densities, who can significantly contribute to the infectious reservoir, is increasing. This poses a limitation to studies relying on the experimental infection of large numbers of mosquitoes with natural isolates of Plasmodium. A simple, field-applicable method is presented for improving parasite infectivity by concentrating Plasmodium falciparum gametocytes. METHODS: Anopheles gambiae received one of the following 5 blood treatments through DMFA: (i) whole blood (WB) samples from naturally-infected donors; (ii) donor blood whose plasma was replaced with the same volume of Plasmodium-naive AB + serum (1:1 control); (iii) plasma replaced with a volume of malaria-naïve AB + serum equivalent to half (1:1/2), or to a quarter (1:1/4), of the initial plasma volume; and (v) donor blood whose plasma was fully removed (RBC). The experiment was repeated 4 times using 4 distinct wild parasite isolates. Seven days post-infection, a total of 1,095 midguts were examined for oocyst presence. RESULTS: Substituting plasma with reduced amounts (1:1/2 and 1:1/4) of Plasmodium-naive AB + serum led to a 31% and 17% increase of the mosquito infection rate and to a 85% and 308% increase in infection intensity compared to the 1:1 control, respectively. The full removal of plasma (RBC) reduced the infection rate by 58% and the intensity by 64% compared to the 1:1 control. Reducing serum volumes (1:1/2; 1:1/4 and RBC) had no impact on mosquito feeding rate and survival when compared to the 1:1 control. CONCLUSIONS: Concentrating gametocytic blood by replacing natural plasma by lower amount of naive serum can enhance the success of mosquito infection. In an area with low gametocyte density, this simple and practical method of parasite concentration can facilitate studies on human-to-mosquito transmission such as the evaluation of transmission-blocking interventions.


Asunto(s)
Anopheles , Mosquitos Vectores , Plasmodium falciparum , Plasmodium falciparum/fisiología , Animales , Anopheles/parasitología , Mosquitos Vectores/parasitología , Humanos , Malaria Falciparum/parasitología , Malaria Falciparum/transmisión , Femenino , Conducta Alimentaria
3.
Trends Parasitol ; 40(5): 416-426, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38637184

RESUMEN

The micropore, a mysterious structure found in apicomplexan species, was recently shown to be essential for nutrient acquisition in Plasmodium falciparum and Toxoplasma gondii. However, the differences between the micropores of these two parasites questions the nature of a general apicomplexan micropore structure and whether the formation process model from Plasmodium can be applied to other apicomplexans. We analyzed the literature on different apicomplexan micropores and found that T. gondii probably harbors a more representative micropore type than the more widely studied ones in Plasmodium. Using recent knowledge of the Kelch 13 (K13) protein interactome and gene depletion phenotypes in the T. gondii micropore, we propose a model of micropore formation, thus enriching our wider understanding of micropore protein function.


Asunto(s)
Apicomplexa , Plasmodium falciparum , Toxoplasma , Apicomplexa/fisiología , Apicomplexa/genética , Toxoplasma/genética , Toxoplasma/fisiología , Plasmodium falciparum/fisiología , Plasmodium falciparum/genética , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/genética
4.
Nat Commun ; 15(1): 3230, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38649361

RESUMEN

Despite concern that climate change could increase the human risk to malaria in certain areas, the temperature dependency of malaria transmission is poorly characterized. Here, we use a mechanistic model fitted to experimental data to describe how Plasmodium falciparum infection of the African malaria vector, Anopheles gambiae, is modulated by temperature, including its influences on parasite establishment, conversion efficiency through parasite developmental stages, parasite development rate, and overall vector competence. We use these data, together with estimates of the survival of infected blood-fed mosquitoes, to explore the theoretical influence of temperature on transmission in four locations in Kenya, considering recent conditions and future climate change. Results provide insights into factors limiting transmission in cooler environments and indicate that increases in malaria transmission due to climate warming in areas like the Kenyan Highlands, might be less than previously predicted.


Asunto(s)
Anopheles , Malaria Falciparum , Mosquitos Vectores , Plasmodium falciparum , Temperatura , Plasmodium falciparum/fisiología , Malaria Falciparum/transmisión , Malaria Falciparum/parasitología , Malaria Falciparum/epidemiología , Animales , Anopheles/parasitología , Humanos , Kenia/epidemiología , Mosquitos Vectores/parasitología , Cambio Climático , Femenino
5.
mBio ; 15(5): e0285023, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38564676

RESUMEN

Condensin I is a pentameric complex that regulates the mitotic chromosome assembly in eukaryotes. The kleisin subunit CAP-H of the condensin I complex acts as a linchpin to maintain the structural integrity and loading of this complex on mitotic chromosomes. This complex is present in all eukaryotes and has recently been identified in Plasmodium spp. However, how this complex is assembled and whether the kleisin subunit is critical for this complex in these parasites are yet to be explored. To examine the role of PfCAP-H during cell division within erythrocytes, we generated an inducible PfCAP-H knockout parasite. We find that PfCAP-H is dynamically expressed during mitosis with the peak expression at the metaphase plate. PfCAP-H interacts with PfCAP-G and is a non-SMC member of the condensin I complex. Notably, the absence of PfCAP-H does not alter the expression of PfCAP-G but affects its localization at the mitotic chromosomes. While mitotic spindle assembly is intact in PfCAP-H-deficient parasites, duplicated centrosomes remain clustered over the mass of unsegmented nuclei with failed karyokinesis. This failure leads to the formation of an abnormal nuclear mass, while cytokinesis occurs normally. Altogether, our data suggest that PfCAP-H plays a crucial role in maintaining the structural integrity of the condensin I complex on the mitotic chromosomes and is essential for the asexual development of malarial parasites. IMPORTANCE: Mitosis is a fundamental process for Plasmodium parasites, which plays a vital role in their survival within two distinct hosts-human and Anopheles mosquitoes. Despite its great significance, our comprehension of mitosis and its regulation remains limited. In eukaryotes, mitosis is regulated by one of the pivotal complexes known as condensin complexes. The condensin complexes are responsible for chromosome condensation, ensuring the faithful distribution of genetic material to daughter cells. While condensin complexes have recently been identified in Plasmodium spp., our understanding of how this complex is assembled and its precise functions during the blood stage development of Plasmodium falciparum remains largely unexplored. In this study, we investigate the role of a central protein, PfCAP-H, during the blood stage development of P. falciparum. Our findings reveal that PfCAP-H is essential and plays a pivotal role in upholding the structure of condensin I and facilitating karyokinesis.


Asunto(s)
Adenosina Trifosfatasas , Proteínas de Unión al ADN , Mitosis , Complejos Multiproteicos , Plasmodium falciparum , Proteínas Protozoarias , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/genética , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Plasmodium falciparum/fisiología , Plasmodium falciparum/crecimiento & desarrollo , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/genética , Eritrocitos/parasitología , Técnicas de Inactivación de Genes , Humanos
6.
Trends Parasitol ; 40(1): 28-44, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38065791

RESUMEN

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.


Asunto(s)
Malaria Cerebral , Humanos , Encéfalo , Plasmodium falciparum/fisiología , Interacciones Huésped-Parásitos , Eritrocitos/parasitología
7.
Brain Res ; 1822: 148669, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37951562

RESUMEN

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.


Asunto(s)
Barrera Hematoencefálica , Bradiquinina , Adhesión Celular , Malaria Cerebral , Malaria Falciparum , Plasmodium falciparum , Humanos , Bradiquinina/metabolismo , Adhesión Celular/fisiología , Medios de Cultivo Condicionados/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Eritrocitos/parasitología , Malaria Cerebral/metabolismo , Malaria Cerebral/parasitología , Malaria Falciparum/metabolismo , Malaria Falciparum/parasitología , Monocitos/fisiología , Plasmodium falciparum/fisiología , Barrera Hematoencefálica/fisiopatología
8.
PLoS Pathog ; 19(12): e1011807, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38051755

RESUMEN

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.


Asunto(s)
Malaria Falciparum , Malaria , Parásitos , Animales , Humanos , Parásitos/fisiología , Malaria Falciparum/parasitología , Malaria/parasitología , Plasmodium falciparum/fisiología , Merozoítos/fisiología , Eritrocitos/parasitología
9.
PLoS Pathog ; 19(10): e1011661, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37856470

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades Intestinales , Malaria Falciparum , Malaria , Humanos , Animales , Ratones , Microcirculación , Malaria Falciparum/parasitología , Malaria/parasitología , Plasmodium falciparum/fisiología
10.
Elife ; 122023 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-37796723

RESUMEN

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.


Asunto(s)
Malaria Falciparum , Plasmodium falciparum , Humanos , Plasmodium falciparum/fisiología , Basigina , Eritrocitos/parasitología , Anticuerpos Neutralizantes , Malaria Falciparum/parasitología , Proteínas Protozoarias/metabolismo , Unión Proteica , Antígenos de Protozoos
11.
Science ; 381(6657): 533-540, 2023 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-37535741

RESUMEN

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.


Asunto(s)
Anopheles , Delftia , Interacciones Huésped-Parásitos , Malaria Falciparum , Plasmodium falciparum , Animales , Anopheles/microbiología , Malaria Falciparum/microbiología , Malaria Falciparum/prevención & control , Malaria Falciparum/transmisión , Plasmodium falciparum/microbiología , Plasmodium falciparum/fisiología , Delftia/fisiología , Simbiosis , Humanos
12.
J Biol Chem ; 299(9): 105111, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37517694

RESUMEN

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.


Asunto(s)
Antimaláricos , Eritrocitos , Glucólisis , Malaria Falciparum , Modelos Biológicos , Terapia Molecular Dirigida , Plasmodium falciparum , Humanos , Acidosis Láctica , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Antimaláricos/metabolismo , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Eritrocitos/parasitología , Glucosa/metabolismo , Glucólisis/efectos de los fármacos , Hipoglucemia , Cinética , Malaria Falciparum/metabolismo , Malaria Falciparum/parasitología , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidad , Plasmodium falciparum/fisiología , Trofozoítos/patogenicidad , Trofozoítos/fisiología , Terapia Molecular Dirigida/métodos , Carga de Parásitos
13.
PLoS Pathog ; 19(6): e1011448, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37339122

RESUMEN

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.


Asunto(s)
Anopheles , Insecticidas , Malaria Falciparum , Malaria , Animales , Femenino , Insecticidas/farmacología , Plasmodium falciparum/fisiología , Resistencia a los Insecticidas/genética , Anopheles/fisiología , Mosquitos Vectores/genética , Lípidos , Control de Mosquitos
14.
Elife ; 122023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36916164

RESUMEN

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.


Asunto(s)
Malaria Falciparum , Malaria , Parásitos , Animales , Niño , Humanos , Plasmodium falciparum/fisiología , Malaria/parasitología , Reproducción , Adaptación Fisiológica , Malaria Falciparum/parasitología
15.
Parasitol Res ; 122(3): 729-737, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36694092

RESUMEN

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.


Asunto(s)
Células Endoteliales , Malaria Cerebral , Humanos , Animales , Ratones , Encéfalo/metabolismo , Barrera Hematoencefálica , Malaria Cerebral/tratamiento farmacológico , Malaria Cerebral/metabolismo , Plasmodium falciparum/fisiología
16.
Microbes Infect ; 25(5): 105102, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36708871

RESUMEN

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.


Asunto(s)
Malaria Falciparum , Plasmodium falciparum , Animales , Plasmodium falciparum/fisiología , Hidrolasas Diéster Fosfóricas , Malaria Falciparum/parasitología , Eritrocitos/parasitología , Transducción de Señal
17.
Parasit Vectors ; 15(1): 434, 2022 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-36397133

RESUMEN

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.


Asunto(s)
Babesia microti , Babesia , Malaria Falciparum , Humanos , Plasmodium falciparum/fisiología , Eritrocitos/parasitología
18.
Adv Sci (Weinh) ; 9(36): e2202944, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36300890

RESUMEN

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.


Asunto(s)
Malaria Cerebral , Malaria Falciparum , Humanos , Malaria Cerebral/parasitología , Malaria Cerebral/patología , Plasmodium falciparum/fisiología , Malaria Falciparum/metabolismo , Malaria Falciparum/parasitología , Encéfalo/patología , Eritrocitos/metabolismo
19.
mBio ; 13(5): e0093722, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-35972144

RESUMEN

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.


Asunto(s)
Antimaláricos , Artemisininas , Plasmodium falciparum , Antimaláricos/farmacología , Artemisininas/farmacología , Mezclas Complejas , Resistencia a Medicamentos/genética , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/fisiología , Proteínas Protozoarias/genética , Aptitud Genética
20.
Methods Mol Biol ; 2470: 493-503, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35881369

RESUMEN

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.


Asunto(s)
Malaria Falciparum , Parásitos , Animales , Niño , Preescolar , Eritrocitos/metabolismo , Citometría de Flujo , Humanos , Malaria Falciparum/parasitología , Plasmodium falciparum/fisiología , Formación de Roseta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA