Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.449
Filter
1.
PLoS One ; 19(7): e0306289, 2024.
Article in English | MEDLINE | ID: mdl-38950022

ABSTRACT

Although the overall burden of malaria is decreasing in Ethiopia, a recent report of an unpredictable increased incidence may be related to the presence of community-wide gametocyte-carrier individuals and a high proportion of infected vectors. This study aimed to reveal the current prevalence of gametocyte-carriage and the sporozoite infectivity rate of Anopheles vectors for Plasmodium parasites. A community-based cross-sectional study was conducted from May 01 to June 30/2019. A total of 53 households were selected using systematic random sampling and a 242 study participants were recruited. Additionally,515 adult female Anopheles mosquitoes were collected using Center for Diseases Control and Prevention (CDC) light traps and mouth aspirators. Parasite gametocytemia was determined using giemsa stain microscopy, while sporozoite infection was determined by giemsa staining microscopy and enzyme linked immunosorbent assay (ELISA). Among the total 242 study participants, 5.4% (95%, CI = 2.9-8.3) of them were positive for any of the Plasmodium species gametocyte. Furthermore, being female [AOR = 15.5(95%, CI = 1.71-140.39)], age group between 15-29 years old [AOR = 16.914 (95%, CI = 1.781-160.63)], no ITNs utilization [AOR = 16.7(95%, CI = 1.902 -146.727)], and high asexual parasite density [(95%, CI = 0.057-0.176, P = 0.001, F = 18.402)] were identified as statistically significant factors for gametocyte carriage. Whereas sporozoite infection rate was 11.6% (95%, CI = 8.2-15.5) and 12.7% (95%, CI = 9.6-16.3) by microscopy and ELISA, respectively. Overall, this study indicated that malaria remains to be an important public health problem in Gondar Zuria district where high gametocyte carriage rate and sporozoite infection rate could sustain its transmission and burden. Therefore, in Ethiopia, where malaria elimination program is underway, frequent, and active community-based surveillance of gametocytemia and sporozoite infection rate is important.


Subject(s)
Anopheles , Mosquito Vectors , Sporozoites , Animals , Ethiopia/epidemiology , Humans , Anopheles/parasitology , Female , Adult , Sporozoites/physiology , Adolescent , Young Adult , Male , Cross-Sectional Studies , Mosquito Vectors/parasitology , Child , Child, Preschool , Malaria/epidemiology , Malaria/parasitology , Malaria/transmission , Middle Aged , Plasmodium/isolation & purification , Infant , Plasmodium falciparum/isolation & purification , Plasmodium falciparum/physiology , Prevalence
2.
Parasit Vectors ; 17(1): 290, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38971776

ABSTRACT

BACKGROUND: Aedes and Anopheles mosquitoes are responsible for tremendous global health burdens from their transmission of pathogens causing malaria, lymphatic filariasis, dengue, and yellow fever. Innovative vector control strategies will help to reduce the prevalence of these diseases. Mass rearing of mosquitoes for research and support of these strategies presently depends on meals of vertebrate blood, which is subject to acquisition, handling, and storage issues. Various blood-free replacements have been formulated for these mosquitoes, but none of these replacements are in wide use, and little is known about their potential impact on competence of the mosquitoes for Plasmodium infection. METHODS: Colonies of Aedes aegypti and Anopheles stephensi were continuously maintained on a blood-free replacement (SkitoSnack; SS) or bovine blood (BB) and monitored for engorgement and hatch rates. Infections of Ae. aegypti and An. stephensi were assessed with Plasmodium gallinaceum and P. falciparum, respectively. RESULTS: Replicate colonies of mosquitoes were maintained on BB or SS for 10 generations of Ae. aegypti and more than 63 generations of An. stephensi. The odds of engorgement by SS- relative to BB-maintained mosquitoes were higher for both Ae. aegypti (OR = 2.6, 95% CI 1.3-5.2) and An. stephensi (OR 2.7, 95% CI 1.4-5.5), while lower odds of hatching were found for eggs from the SS-maintained mosquitoes of both species (Ae. aegypti OR = 0.40, 95% CI 0.26-0.62; An. stephensi OR = 0.59, 95% CI 0.36-0.96). Oocyst counts were similar for P. gallinaceum infections of Ae. aegypti mosquitoes maintained on SS or BB (mean ratio = [mean on SS]/[mean on BB] = 1.11, 95% CI 0.85-1.49). Similar oocyst counts were also observed from the P. falciparum infections of SS- or BB-maintained An. stephensi (mean ratio = 0.76, 95% CI 0.44-1.37). The average counts of sporozoites/mosquito showed no evidence of reductions in the SS-maintained relative to BB-maintained mosquitoes of both species. CONCLUSIONS: Aedes aegypti and An. stephensi can be reliably maintained on SS over multiple generations and are as competent for Plasmodium infection as mosquitoes maintained on BB. Use of SS alleviates the need to acquire and preserve blood for mosquito husbandry and may support new initiatives in fundamental and applied research, including novel manipulations of midgut microbiota and factors important to the mosquito life cycle and pathogen susceptibility.


Subject(s)
Aedes , Anopheles , Mosquito Vectors , Animals , Aedes/parasitology , Aedes/physiology , Anopheles/parasitology , Anopheles/physiology , Mosquito Vectors/parasitology , Mosquito Vectors/physiology , Plasmodium gallinaceum/physiology , Plasmodium falciparum/physiology , Cattle , Female , Blood/parasitology , Feeding Behavior
3.
Nat Commun ; 15(1): 5794, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987258

ABSTRACT

Plasmodium falciparum is the causative agent of malaria and remains a pathogen of global importance. Asexual blood stage replication, via a process called schizogony, is an important target for the development of new antimalarials. Here we use ultrastructure-expansion microscopy to probe the organisation of the chromosome-capturing kinetochores in relation to the mitotic spindle, the centriolar plaque, the centromeres and the apical organelles during schizont development. Conditional disruption of the kinetochore components, PfNDC80 and PfNuf2, is associated with aberrant mitotic spindle organisation, disruption of the centromere marker, CENH3 and impaired karyokinesis. Surprisingly, kinetochore disruption also leads to disengagement of the centrosome equivalent from the nuclear envelope. Severing the connection between the nucleus and the apical complex leads to the formation of merozoites lacking nuclei. Here, we show that correct assembly of the kinetochore/spindle complex plays a previously unrecognised role in positioning the nascent apical complex in developing P. falciparum merozoites.


Subject(s)
Centrosome , Kinetochores , Plasmodium falciparum , Protozoan Proteins , Spindle Apparatus , Kinetochores/metabolism , Plasmodium falciparum/metabolism , Plasmodium falciparum/physiology , Centrosome/metabolism , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Spindle Apparatus/metabolism , Humans , Merozoites/metabolism , Merozoites/physiology , Mitosis , Centromere/metabolism , Nuclear Envelope/metabolism , Malaria, Falciparum/parasitology , Malaria, Falciparum/metabolism
4.
Front Cell Infect Microbiol ; 14: 1408451, 2024.
Article in English | MEDLINE | ID: mdl-38828264

ABSTRACT

Recent studies indicate that human spleen contains over 95% of the total parasite biomass during chronic asymptomatic infections caused by Plasmodium vivax. Previous studies have demonstrated that extracellular vesicles (EVs) secreted from infected reticulocytes facilitate binding to human spleen fibroblasts (hSFs) and identified parasite genes whose expression was dependent on an intact spleen. Here, we characterize the P. vivax spleen-dependent hypothetical gene (PVX_114580). Using CRISPR/Cas9, PVX_114580 was integrated into P. falciparum 3D7 genome and expressed during asexual stages. Immunofluorescence analysis demonstrated that the protein, which we named P. vivax Spleen-Dependent Protein 1 (PvSDP1), was located at the surface of infected red blood cells in the transgenic line and this localization was later confirmed in natural infections. Plasma-derived EVs from P. vivax-infected individuals (PvEVs) significantly increased cytoadherence of 3D7_PvSDP1 transgenic line to hSFs and this binding was inhibited by anti-PvSDP1 antibodies. Single-cell RNAseq of PvEVs-treated hSFs revealed increased expression of adhesion-related genes. These findings demonstrate the importance of parasite spleen-dependent genes and EVs from natural infections in the formation of intrasplenic niches in P. vivax, a major challenge for malaria elimination.


Subject(s)
Extracellular Vesicles , Malaria, Vivax , Plasmodium vivax , Protozoan Proteins , Spleen , Extracellular Vesicles/metabolism , Plasmodium vivax/genetics , Plasmodium vivax/metabolism , Humans , Spleen/metabolism , Spleen/parasitology , Malaria, Vivax/parasitology , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Erythrocytes/parasitology , Erythrocytes/metabolism , Fibroblasts/parasitology , Fibroblasts/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Plasmodium falciparum/physiology , Cell Adhesion , Host-Parasite Interactions
5.
Zhongguo Xue Xi Chong Bing Fang Zhi Za Zhi ; 36(2): 219-220, 2024 May 08.
Article in Chinese | MEDLINE | ID: mdl-38857970

ABSTRACT

Plasmodium falciparum malaria, caused by Plasmodium falciparum infection, is an Anopheles mosquito-transmitted infectious diseases, which predominantly occurs in tropical areas of Africa. P. falciparum malaria is characterized by complex and atypical clinical manifestations, and high likelihood of misdiagnosis and missing diagnosis, and may be life-threatening if treated untimely. This case report presents the diagnosis and treatment of a P. falciparum malaria case with acute abdominal pain as the first symptom.


Subject(s)
Abdominal Pain , Malaria, Falciparum , Humans , Malaria, Falciparum/diagnosis , Malaria, Falciparum/complications , Abdominal Pain/etiology , Abdominal Pain/parasitology , Male , Plasmodium falciparum/isolation & purification , Plasmodium falciparum/physiology , Adult
6.
Mol Biochem Parasitol ; 259: 111634, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38823647

ABSTRACT

Asexual blood stage culture of Plasmodium falciparum is routinely performed but reproducibly inducing commitment to and maturation of viable gametocytes remains difficult. Culture media can be supplemented with human serum substitutes to induce commitment but these generally only allow for long-term culture of asexual parasites and not transmission-competent gametocytes due to their different lipid composition. Recent insights demonstrated the important roles lipids play in sexual commitment; elaborating on this we exposed ring stage parasites (20-24 hours hpi) for one day to AlbuMAX supplemented media to trigger induction to gametocytogenesis. We observed a significant increase in gametocytes after AlbuMAX induction compared to serum. We also tested the transmission potential of AlbuMAX inducted gametocytes and found a significant higher oocyst intensity compared to serum. We conclude that AlbuMAX supplemented media induces commitment, allows a more stable and predictable production of transmittable gametocytes than serum alone.


Subject(s)
Culture Media , Plasmodium falciparum , Plasmodium falciparum/drug effects , Plasmodium falciparum/growth & development , Plasmodium falciparum/physiology , Culture Media/chemistry , Humans , Malaria, Falciparum/parasitology , Malaria, Falciparum/transmission
7.
PLoS Pathog ; 20(6): e1012265, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38829893

ABSTRACT

Asexual replication of Plasmodium falciparum occurs via schizogony, wherein 16-36 daughter cells are produced within the parasite during one semi-synchronized cytokinetic event. Schizogony requires a divergent contractile ring structure known as the basal complex. Our lab has previously identified PfMyoJ (PF3D7_1229800) and PfSLACR (PF3D7_0214700) as basal complex proteins recruited midway through segmentation. Using ultrastructure expansion microscopy, we localized both proteins to a novel basal complex subcompartment. While both colocalize with the basal complex protein PfCINCH upon recruitment, they form a separate, more basal subcompartment termed the posterior cup during contraction. We also show that PfSLACR is recruited to the basal complex prior to PfMyoJ, and that both proteins are removed unevenly as segmentation concludes. Using live-cell microscopy, we show that actin dynamics are dispensable for basal complex formation, expansion, and contraction. We then show that EF-hand containing P. falciparum Centrin 2 partially localizes to this posterior cup of the basal complex and that it is essential for growth and replication, with variable defects in basal complex contraction and synchrony. Finally, we demonstrate that free intracellular calcium is necessary but not sufficient for basal complex contraction in P. falciparum. Thus, we demonstrate dynamic spatial compartmentalization of the Plasmodium falciparum basal complex, identify an additional basal complex protein, and begin to elucidate the unique mechanism of contraction utilized by P. falciparum, opening the door for further exploration of Apicomplexan cellular division.


Subject(s)
Plasmodium falciparum , Protozoan Proteins , Plasmodium falciparum/physiology , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Malaria, Falciparum/parasitology , Malaria, Falciparum/metabolism , Humans , Erythrocytes/parasitology
8.
Microbiol Spectr ; 12(7): e0072724, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38864616

ABSTRACT

A hallmark of cerebral malaria (CM) is sequestration of Plasmodium falciparum-infected erythrocytes (IE) within the brain microvasculature. Binding of IE to endothelium reduces microvascular flow and, combined with an inflammatory response, perturbs endothelial barrier function, resulting in breakdown of the blood-brain barrier (BBB). Cytoadherence leads to activation of the endothelium and alters a range of cell processes affecting signaling pathways, receptor expression, coagulation, and disruption of BBB integrity. Here, we investigated whether CM-derived parasites elicit differential effects on human brain microvascular endothelial cells (HBMECs), as compared to uncomplicated malaria (UM)-derived parasites. Patient-derived IE from UM and CM clinical cases, as well as non-binding skeleton-binding protein 1 knockout parasites, were overlaid onto tumour necrosis factor (TNF)-activated HBMECs. Gene expression analysis of endothelial responses was performed using probe-based assays of a panel of genes involved in inflammation, apoptosis, endothelial barrier function, and prostacyclin synthesis pathway. We observed a significant effect on endothelial transcriptional responses in the presence of IE, yet there was no significant correlation between HBMEC responses and type of clinical syndrome (UM or CM). Furthermore, there was no correlation between HBMEC gene expression and both binding itself and level of IE binding to HBMECs, as we detected the same change in endothelial responses when employing both binding and non-binding parasites. Our results suggest that interaction of IE with endothelial cells in this co-culture model induces some endothelial responses that are independent of clinical origin and independent of the expression of the major variant antigen Plasmodium falciparum erythrocyte membrane protein 1 on the IE surface. IMPORTANCE: Cerebral malaria (CM) is the most prevalent and deadly complication of severe Plasmodium falciparum infection. A hallmark of this disease is sequestration of P. falciparum-infected erythrocytes (IE) in brain microvasculature that ultimately results in breakdown of the blood-brain barrier. Here, we compared the effect of P. falciparum parasites derived from uncomplicated malaria (UM) and CM cases on the relative gene expression of human brain microvascular endothelial cells (HBMECs) for a panel of genes. We observed a significant effect on the endothelial transcriptional response in the presence of IE, yet there is no significant correlation between HBMEC responses and the type of clinical syndrome (UM or CM). Furthermore, there was no correlation between HBMEC gene expression and both binding itself and the level of IE binding to HBMECs. Our results suggest that interaction of IE with endothelial cells induces endothelial responses that are independent of clinical origin and not entirely driven by surface Plasmodium falciparum erythrocyte membrane protein 1 expression.


Subject(s)
Blood-Brain Barrier , Brain , Endothelial Cells , Erythrocytes , Malaria, Cerebral , Malaria, Falciparum , Plasmodium falciparum , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Plasmodium falciparum/physiology , Humans , Endothelial Cells/parasitology , Endothelial Cells/metabolism , Malaria, Falciparum/parasitology , Malaria, Falciparum/metabolism , Malaria, Cerebral/parasitology , Malaria, Cerebral/metabolism , Brain/parasitology , Brain/metabolism , Blood-Brain Barrier/parasitology , Blood-Brain Barrier/metabolism , Erythrocytes/parasitology , Erythrocytes/metabolism
9.
Malar J ; 23(1): 189, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38880891

ABSTRACT

BACKGROUND: Malaria, a prominent vector borne disease causing over a million annual cases worldwide, predominantly affects vulnerable populations in the least developed regions. Despite their preventable and treatable nature, malaria remains a global public health concern. In the last decade, India has faced a significant decline in malaria morbidity and mortality. As India pledged to eliminate malaria by 2030, this study examined a decade of surveillance data to uncover space-time clustering and seasonal trends of Plasmodium vivax and Plasmodium falciparum malaria cases in West Bengal. METHODS: Seasonal and trend decomposition using Loess (STL) was applied to detect seasonal trend and anomaly of the time series. Univariate and multivariate space-time cluster analysis of both malaria cases were performed at block level using Kulldorff's space-time scan statistics from April 2011 to March 2021 to detect statistically significant space-time clusters. RESULTS: From the time series decomposition, a clear seasonal pattern is visible for both malaria cases. Statistical analysis indicated considerable high-risk P. vivax clusters, particularly in the northern, central, and lower Gangetic areas. Whereas, P. falciparum was concentrated in the western region with a significant recent transmission towards the lower Gangetic plain. From the multivariate space-time scan statistics, the co-occurrence of both cases were detected with four significant clusters, which signifies the regions experiencing a greater burden of malaria cases. CONCLUSIONS: Seasonal trends from the time series decomposition analysis show a gradual decline for both P. vivax and P. falciparum cases in West Bengal. The space-time scan statistics identified high-risk blocks for P. vivax and P. falciparum malaria and its co-occurrence. Both malaria types exhibit significant spatiotemporal variations over the study area. Identifying emerging high-risk areas of P. falciparum malaria over the Gangetic belt indicates the need for more research for its spatial shifting. Addressing the drivers of malaria transmission in these diverse clusters demands regional cooperation and strategic strategies, crucial steps towards overcoming the final obstacles in malaria eradication.


Subject(s)
Malaria, Falciparum , Malaria, Vivax , Plasmodium vivax , Seasons , India/epidemiology , Malaria, Vivax/epidemiology , Malaria, Vivax/parasitology , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Humans , Plasmodium vivax/physiology , Space-Time Clustering , Plasmodium falciparum/physiology
10.
Malar J ; 23(1): 130, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693572

ABSTRACT

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.


Subject(s)
Chorionic Villi , Plasmodium falciparum , Trophoblasts , Humans , Female , Chorionic Villi/parasitology , Chorionic Villi/pathology , Pregnancy , Plasmodium falciparum/physiology , Trophoblasts/parasitology , Apoptosis , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Placenta/parasitology , Placenta/pathology , Cytokines/metabolism
11.
Parasit Vectors ; 17(1): 227, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755646

ABSTRACT

Volatile organic compounds (VOCs) are chemicals emitted as products of cell metabolism, which reflects the physiological and pathological conditions of any living organisms. These compounds play a key role as olfactory cues for arthropod vectors such as mosquitoes, sand flies, and ticks, which act in the transmission of pathogens to many animal species, including humans. Some VOCs may influence arthropod behaviour, e.g., host preference and oviposition site selection for gravid females. Furthermore, deadly vector-borne pathogens such as Plasmodium falciparum and Leishmania infantum are suggested to manipulate the VOCs profile of the host to make them more attractive to mosquitoes and sand fly vectors, respectively. Under the above circumstances, studies on these compounds have demonstrated their potential usefulness for investigating the behavioural response of mosquitoes, sand flies, and ticks toward their vertebrate hosts, as well as potential tools for diagnosis of vector-borne diseases (VBDs). Herein, we provide an account for scientific data available on VOCs to study the host seeking behaviour of arthropod vectors, and their usefulness as attractants, repellents, or tools for an early diagnosis of VBDs.


Subject(s)
Culicidae , Psychodidae , Ticks , Volatile Organic Compounds , Animals , Volatile Organic Compounds/metabolism , Psychodidae/physiology , Psychodidae/parasitology , Ticks/physiology , Humans , Culicidae/physiology , Behavior, Animal , Vector Borne Diseases/transmission , Female , Mosquito Vectors/physiology , Mosquito Vectors/parasitology , Plasmodium falciparum/physiology
12.
Malar J ; 23(1): 135, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711028

ABSTRACT

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.


Subject(s)
Anopheles , Mosquito Vectors , Plasmodium falciparum , Plasmodium falciparum/physiology , Animals , Anopheles/parasitology , Mosquito Vectors/parasitology , Humans , Malaria, Falciparum/parasitology , Malaria, Falciparum/transmission , Female , Feeding Behavior
13.
Sensors (Basel) ; 24(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38794040

ABSTRACT

Malaria is a disease that affects millions of people worldwide, particularly in developing countries. The development of accurate and efficient methods for the detection of malaria-infected cells is crucial for effective disease management and control. This paper presents the electrical impedance spectroscopy (EIS) of normal and malaria-infected red blood cells. An EIS microfluidic device, comprising a microchannel and a pair of coplanar electrodes, was fabricated for single-cell measurements in a continuous manner. Based on the EIS results, the aim of this work is to discriminate Plasmodium falciparum-infected red blood cells from the normal ones. Different from typical impedance spectroscopy, our measurement was performed for the cells in a low-conductivity medium in a frequency range between 50 kHz and 800 kHz. Numerical simulation was utilized to study the suitability parameters of the microchannel and electrodes for the EIS experiment over the measurement frequencies. The measurement results have shown that by using the low-conductivity medium, we could focus on the change in the conductance caused by the presence of a cell in the sensing electrode gap. The results indicated a distinct frequency spectrum of the conductance between the normal and infected red blood cells, which can be further used for the detection of the disease.


Subject(s)
Dielectric Spectroscopy , Erythrocytes , Plasmodium falciparum , Erythrocytes/parasitology , Dielectric Spectroscopy/methods , Dielectric Spectroscopy/instrumentation , Humans , Plasmodium falciparum/physiology , Plasmodium falciparum/pathogenicity , Electrodes , Lab-On-A-Chip Devices , Malaria, Falciparum/diagnosis , Malaria, Falciparum/parasitology , Electric Impedance , Malaria/diagnosis , Malaria/parasitology
14.
Nat Commun ; 15(1): 4626, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816383

ABSTRACT

The human infectious reservoir of Plasmodium falciparum is governed by transmission efficiency during vector-human contact and mosquito biting preferences. Understanding biting bias in a natural setting can help target interventions to interrupt transmission. In a 15-month cohort in western Kenya, we detected P. falciparum in indoor-resting Anopheles and human blood samples by qPCR and matched mosquito bloodmeals to cohort participants using short-tandem repeat genotyping. Using risk factor analyses and discrete choice models, we assessed mosquito biting behavior with respect to parasite transmission. Biting was highly unequal; 20% of people received 86% of bites. Biting rates were higher on males (biting rate ratio (BRR): 1.68; CI: 1.28-2.19), children 5-15 years (BRR: 1.49; CI: 1.13-1.98), and P. falciparum-infected individuals (BRR: 1.25; CI: 1.01-1.55). In aggregate, P. falciparum-infected school-age (5-15 years) boys accounted for 50% of bites potentially leading to onward transmission and had an entomological inoculation rate 6.4x higher than any other group. Additionally, infectious mosquitoes were nearly 3x more likely than non-infectious mosquitoes to bite P. falciparum-infected individuals (relative risk ratio 2.76, 95% CI 1.65-4.61). Thus, persistent P. falciparum transmission was characterized by disproportionate onward transmission from school-age boys and by the preference of infected mosquitoes to feed upon infected people.


Subject(s)
Anopheles , Insect Bites and Stings , Malaria, Falciparum , Mosquito Vectors , Plasmodium falciparum , Humans , Anopheles/parasitology , Anopheles/physiology , Animals , Plasmodium falciparum/physiology , Plasmodium falciparum/isolation & purification , Plasmodium falciparum/genetics , Malaria, Falciparum/transmission , Malaria, Falciparum/parasitology , Male , Adolescent , Child , Child, Preschool , Female , Kenya/epidemiology , Mosquito Vectors/parasitology , Mosquito Vectors/physiology , Adult , Feeding Behavior , Young Adult , Infant
15.
Nat Commun ; 15(1): 3230, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649361

ABSTRACT

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.


Subject(s)
Anopheles , Malaria, Falciparum , Mosquito Vectors , Plasmodium falciparum , Temperature , Plasmodium falciparum/physiology , Malaria, Falciparum/transmission , Malaria, Falciparum/parasitology , Malaria, Falciparum/epidemiology , Animals , Anopheles/parasitology , Humans , Kenya/epidemiology , Mosquito Vectors/parasitology , Climate Change , Female
16.
Curr Opin Microbiol ; 79: 102469, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38574448

ABSTRACT

Malaria blood stage parasites commit to either one of two distinct cellular fates while developing within erythrocytes of their mammalian host: they either undergo another round of asexual replication or they differentiate into nonreplicative transmissible gametocytes. Depending on the state of infection, either path may support or impair the ultimate goal of human-to-human transmission via the mosquito vector. Malaria parasites therefore evolved strategies to control investments into asexual proliferation versus gametocyte formation. Recent work provided fascinating molecular insight into shared and unique mechanisms underlying the control and environmental modulation of sexual commitment in the two most widely studied malaria parasite species, Plasmodium falciparum and P. berghei. With this review, we aim at placing these findings into a comparative mechanistic context.


Subject(s)
Plasmodium berghei , Plasmodium falciparum , Plasmodium falciparum/physiology , Plasmodium falciparum/growth & development , Plasmodium falciparum/genetics , Animals , Humans , Plasmodium berghei/physiology , Plasmodium berghei/growth & development , Plasmodium berghei/genetics , Malaria/parasitology , Malaria/transmission , Erythrocytes/parasitology
17.
Trends Parasitol ; 40(5): 416-426, 2024 May.
Article in English | MEDLINE | ID: mdl-38637184

ABSTRACT

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.


Subject(s)
Apicomplexa , Plasmodium falciparum , Toxoplasma , Apicomplexa/physiology , Apicomplexa/genetics , Toxoplasma/genetics , Toxoplasma/physiology , Plasmodium falciparum/physiology , Plasmodium falciparum/genetics , Protozoan Proteins/metabolism , Protozoan Proteins/genetics
18.
mBio ; 15(5): e0285023, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38564676

ABSTRACT

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.


Subject(s)
Adenosine Triphosphatases , Cell Nucleus Division , DNA-Binding Proteins , Mitosis , Plasmodium falciparum , Humans , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Erythrocytes/parasitology , Gene Knockout Techniques , Multiprotein Complexes/metabolism , Multiprotein Complexes/genetics , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Plasmodium falciparum/physiology , Plasmodium falciparum/growth & development , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Cell Nucleus Division/genetics
19.
Trends Parasitol ; 40(1): 28-44, 2024 01.
Article in English | MEDLINE | ID: mdl-38065791

ABSTRACT

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.


Subject(s)
Malaria, Cerebral , Humans , Brain , Plasmodium falciparum/physiology , Host-Parasite Interactions , Erythrocytes/parasitology
20.
Brain Res ; 1822: 148669, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37951562

ABSTRACT

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


Subject(s)
Blood-Brain Barrier , Bradykinin , Cell Adhesion , Malaria, Cerebral , Malaria, Falciparum , Plasmodium falciparum , Humans , Bradykinin/metabolism , Cell Adhesion/physiology , Culture Media, Conditioned/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Erythrocytes/parasitology , Malaria, Cerebral/metabolism , Malaria, Cerebral/parasitology , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Monocytes/physiology , Plasmodium falciparum/physiology , Blood-Brain Barrier/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...