Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.614
Filtrer
1.
PLoS One ; 19(7): e0306289, 2024.
Article de Anglais | MEDLINE | ID: mdl-38950022

RÉSUMÉ

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.


Sujet(s)
Anopheles , Vecteurs moustiques , Sporozoïtes , Animaux , Éthiopie/épidémiologie , Humains , Anopheles/parasitologie , Femelle , Adulte , Sporozoïtes/physiologie , Adolescent , Jeune adulte , Mâle , Études transversales , Vecteurs moustiques/parasitologie , Enfant , Enfant d'âge préscolaire , Paludisme/épidémiologie , Paludisme/parasitologie , Paludisme/transmission , Adulte d'âge moyen , Plasmodium/isolement et purification , Nourrisson , Plasmodium falciparum/isolement et purification , Plasmodium falciparum/physiologie , Prévalence
2.
PLoS One ; 19(7): e0307291, 2024.
Article de Anglais | MEDLINE | ID: mdl-39024284

RÉSUMÉ

Cell culture systems have long been recognised as great resources to mitigate the use of animals in research, offering effective solutions for replacement or reduction with benefits commonly including lower costs, shorter duration and improved reproducibility. The use of in vitro culture methods has been extensively explored for many apicomplexan parasites, supporting significant research advances, but studies with Eimeria are often limited since they still depend on the animal host. In this study we have used 2.5D and 3D culture systems for the first time to evaluate the growth of Eimeria tenella parasites using a panel of cell lines (MDBK, HD11, COLO-680N and HCC4006). Results were compared to growth in 2D monolayers following established protocols. Observations using the fluorescent transgenic strain Et-dYFP showed invasion and development of parasites inside cells suspended in a collagen matrix (2.5D or 3D), supporting the development of asexual stages with the release of first-generation merozoites. Similar findings were observed when Scaffold-free 3D cell spheroids of HD11 cells were infected with sporozoites. No subsequent developmental stages were identified while evaluating these cell lines and further work will be required to improve in vitro culture systems to a point where reduction and replacement of animal use becomes routine.


Sujet(s)
Techniques de culture cellulaire , Eimeria tenella , Eimeria tenella/croissance et développement , Animaux , Techniques de culture cellulaire/méthodes , Humains , Lignée cellulaire , Sporozoïtes/croissance et développement , Sphéroïdes de cellules/parasitologie
3.
Proc Natl Acad Sci U S A ; 121(28): e2403442121, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38968107

RÉSUMÉ

Plasmodium falciparum causes severe malaria and assembles a protein translocon (PTEX) complex at the parasitophorous vacuole membrane (PVM) of infected erythrocytes, through which several hundred proteins are exported to facilitate growth. The preceding liver stage of infection involves growth in a hepatocyte-derived PVM; however, the importance of protein export during P. falciparum liver infection remains unexplored. Here, we use the FlpL/FRT system to conditionally excise genes in P. falciparum sporozoites for functional liver-stage studies. Disruption of PTEX members ptex150 and exp2 did not affect sporozoite development in mosquitoes or infectivity for hepatocytes but attenuated liver-stage growth in humanized mice. While PTEX150 deficiency reduced fitness on day 6 postinfection by 40%, EXP2 deficiency caused 100% loss of liver parasites, demonstrating that PTEX components are required for growth in hepatocytes to differing degrees. To characterize PTEX loss-of-function mutations, we localized four liver-stage Plasmodium export element (PEXEL) proteins. P. falciparum liver specific protein 2 (LISP2), liver-stage antigen 3 (LSA3), circumsporozoite protein (CSP), and a Plasmodium berghei LISP2 reporter all localized to the periphery of P. falciparum liver stages but were not exported beyond the PVM. Expression of LISP2 and CSP but not LSA3 was reduced in ptex150-FRT and exp2-FRT liver stages, suggesting that expression of some PEXEL proteins is affected directly or indirectly by PTEX disruption. These results show that PTEX150 and EXP2 are important for P. falciparum development in hepatocytes and emphasize the emerging complexity of PEXEL protein trafficking.


Sujet(s)
Hépatocytes , Foie , Paludisme à Plasmodium falciparum , Plasmodium falciparum , Protéines de protozoaire , Sporozoïtes , Plasmodium falciparum/croissance et développement , Plasmodium falciparum/génétique , Plasmodium falciparum/métabolisme , Animaux , Protéines de protozoaire/métabolisme , Protéines de protozoaire/génétique , Sporozoïtes/métabolisme , Sporozoïtes/croissance et développement , Souris , Foie/parasitologie , Foie/métabolisme , Humains , Hépatocytes/parasitologie , Hépatocytes/métabolisme , Paludisme à Plasmodium falciparum/parasitologie
4.
Proc Natl Acad Sci U S A ; 121(24): e2320898121, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38833464

RÉSUMÉ

The World Health Organization identifies a strong surveillance system for malaria and its mosquito vector as an essential pillar of the malaria elimination agenda. Anopheles salivary antibodies are emerging biomarkers of exposure to mosquito bites that potentially overcome sensitivity and logistical constraints of traditional entomological surveys. Using samples collected by a village health volunteer network in 104 villages in Southeast Myanmar during routine surveillance, the present study employs a Bayesian geostatistical modeling framework, incorporating climatic and environmental variables together with Anopheles salivary antigen serology, to generate spatially continuous predictive maps of Anopheles biting exposure. Our maps quantify fine-scale spatial and temporal heterogeneity in Anopheles salivary antibody seroprevalence (ranging from 9 to 99%) that serves as a proxy of exposure to Anopheles bites and advances current static maps of only Anopheles occurrence. We also developed an innovative framework to perform surveillance of malaria transmission. By incorporating antibodies against the vector and the transmissible form of malaria (sporozoite) in a joint Bayesian geostatistical model, we predict several foci of ongoing transmission. In our study, we demonstrate that antibodies specific for Anopheles salivary and sporozoite antigens are a logistically feasible metric with which to quantify and characterize heterogeneity in exposure to vector bites and malaria transmission. These approaches could readily be scaled up into existing village health volunteer surveillance networks to identify foci of residual malaria transmission, which could be targeted with supplementary interventions to accelerate progress toward elimination.


Sujet(s)
Anopheles , Théorème de Bayes , Paludisme , Vecteurs moustiques , Animaux , Anopheles/parasitologie , Vecteurs moustiques/parasitologie , Humains , Paludisme/transmission , Paludisme/épidémiologie , Paludisme/immunologie , Paludisme/parasitologie , Études séroépidémiologiques , Morsures et piqûres d'insectes/épidémiologie , Morsures et piqûres d'insectes/immunologie , Morsures et piqûres d'insectes/parasitologie , Sporozoïtes/immunologie
5.
PLoS Genet ; 20(6): e1011162, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38885280

RÉSUMÉ

Very little is known about the process of meiosis in the apicomplexan parasite Cryptosporidium despite the essentiality of sex in its life cycle. Most cell lines only support asexual growth of Cryptosporidium parvum (C. parvum), but stem cell derived intestinal epithelial cells grown under air-liquid interface (ALI) conditions support the sexual cycle. To examine chromosomal dynamics during meiosis in C. parvum, we generated two transgenic lines of parasites that were fluorescently tagged with mCherry or GFP on chromosomes 1 or 5, respectively. Infection of ALI cultures or Ifngr1-/- mice with mCherry and GFP parasites resulted in cross-fertilization and the formation of "yellow" oocysts, which contain 4 haploid sporozoites that are the product of meiosis. Recombinant oocysts from the F1 generation were purified and used to infect HCT-8 cultures, and phenotypes of the progeny were observed by microscopy. All possible phenotypes predicted by independent segregation were represented equally (~25%) in the population, indicating that C. parvum chromosomes exhibit a Mendelian inheritance pattern. The most common pattern observed from the outgrowth of single oocysts included all possible parental and recombinant phenotypes derived from a single meiotic event, suggesting a high rate of crossover. To estimate the frequency of crossover, additional loci on chromosomes 1 and 5 were tagged and used to monitor intrachromosomal crosses in Ifngr1-/- mice. Both chromosomes showed a high frequency of crossover compared to other apicomplexans with map distances (i.e., 1% recombination) of 3-12 kb. Overall, a high recombination rate may explain many unique characteristics observed in Cryptosporidium spp. such as high rates of speciation, wide variation in host range, and rapid evolution of host-specific virulence factors.


Sujet(s)
Cryptosporidiose , Cryptosporidium parvum , Méiose , Oocystes , Recombinaison génétique , Animaux , Cryptosporidium parvum/génétique , Souris , Cryptosporidiose/parasitologie , Cryptosporidiose/génétique , Méiose/génétique , Humains , Récepteur interféron/génétique , , Ségrégation des chromosomes/génétique , Sporozoïtes/génétique , Souris knockout , Phénotype
6.
Sci Rep ; 14(1): 13669, 2024 06 13.
Article de Anglais | MEDLINE | ID: mdl-38871839

RÉSUMÉ

Among the factors affecting the effectiveness of malaria control is poor knowledge of the entomologic drivers of the disease. We investigated anopheline populations as part of a baseline study to implement house screening of windows and doors as a supplementary malaria control tool towards elimination in Jabi Tehnan district, Amhara Regional State of Ethiopia. The samples were surveyed monthly using CDC light traps between June 2020 and May 2021. Mosquito trap density (< 3 mosquitoes/trap) was low, however, with a high overall Plasmodium sporozoite rate (9%; indoor = 4.3%, outdoor = 13.1%) comprising P. falciparum (88.9%) and P. vivax (11.1%). Anopheles gambiae s.l., mostly An. arabiensis, comprised > 80% of total anopheline captures and contributed ~ 42% of Plasmodium-infected mosquitoes. On the other hand, morphologically scored Anopheles funestus s.l., constituting about 6% of anopheline collections, accounted for 50% of sporozoite-infected mosquitoes. Most of the infected An. funestus s.l. specimens (86.7%) were grouped with previously unknown or undescribed Anopheles species previously implicated as a cryptic malaria vector in the western Kenyan highlands, confirming its wider geographic distribution in eastern Africa. Other species with Plasmodium infection included An. longipalpis C, An. theileri, An. demillioni, and An. nili. Cumulatively, 77.8% of the infected mosquitoes occurred outdoors. These results suggest efficient malaria parasite transmission despite the low vector densities, which has implications for effective endpoint indicators to monitor malaria control progress. Additionally, the largely outdoor infection and discovery of previously unknown and cryptic vectors suggest an increased risk of residual malaria transmission and, thus, a constraint on effective malaria prevention and control.


Sujet(s)
Anopheles , Vecteurs moustiques , Éthiopie/épidémiologie , Animaux , Anopheles/parasitologie , Vecteurs moustiques/parasitologie , Humains , Paludisme/transmission , Paludisme/épidémiologie , Plasmodium falciparum/isolement et purification , Plasmodium falciparum/pathogénicité , Plasmodium vivax/physiologie , Sporozoïtes , Lutte contre les moustiques/méthodes , Paludisme à Plasmodium vivax/transmission , Paludisme à Plasmodium vivax/épidémiologie , Paludisme à Plasmodium vivax/parasitologie , Paludisme à Plasmodium falciparum/transmission , Paludisme à Plasmodium falciparum/épidémiologie , Paludisme à Plasmodium falciparum/parasitologie , Femelle
7.
J Cell Sci ; 137(11)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38832798

RÉSUMÉ

Plasmodium sporozoites are the infective forms of the malaria parasite in the mosquito and vertebrate host. Gliding motility allows sporozoites to migrate and invade mosquito salivary glands and mammalian hosts. Motility and invasion are powered by an actin-myosin motor complex linked to the glideosome, which contains glideosome-associated proteins (GAPs), MyoA and the myosin A tail-interacting protein (MTIP). However, the role of several proteins involved in gliding motility remains unknown. We identified that the S14 gene is upregulated in sporozoite from transcriptome data of Plasmodium yoelii and further confirmed its transcription in P. berghei sporozoites using real-time PCR. C-terminal 3×HA-mCherry tagging revealed that S14 is expressed and localized on the inner membrane complex of the sporozoites. We disrupted S14 in P. berghei and demonstrated that it is essential for sporozoite gliding motility, and salivary gland and hepatocyte invasion. The gliding and invasion-deficient S14 knockout sporozoites showed normal expression and organization of inner membrane complex and surface proteins. Taken together, our data show that S14 plays a role in the function of the glideosome and is essential for malaria transmission.


Sujet(s)
Paludisme , Plasmodium berghei , Protéines de protozoaire , Sporozoïtes , Sporozoïtes/métabolisme , Plasmodium berghei/métabolisme , Plasmodium berghei/génétique , Protéines de protozoaire/métabolisme , Protéines de protozoaire/génétique , Animaux , Souris , Paludisme/parasitologie , Glandes salivaires/parasitologie , Glandes salivaires/métabolisme , Anopheles/parasitologie
8.
JCI Insight ; 9(9)2024 May 08.
Article de Anglais | MEDLINE | ID: mdl-38716733

RÉSUMÉ

Vaccination of malaria-naive volunteers with a high dose of Plasmodium falciparum sporozoites chemoattenuated by chloroquine (CQ) (PfSPZ-CVac [CQ]) has previously demonstrated full protection against controlled human malaria infection (CHMI). However, lower doses of PfSPZ-CVac [CQ] resulted in incomplete protection. This provides the opportunity to understand the immune mechanisms needed for better vaccine-induced protection by comparing individuals who were protected with those not protected. Using mass cytometry, we characterized immune cell composition and responses of malaria-naive European volunteers who received either lower doses of PfSPZ-CVac [CQ], resulting in 50% protection irrespective of the dose, or a placebo vaccination, with everyone becoming infected following CHMI. Clusters of CD4+ and γδ T cells associated with protection were identified, consistent with their known role in malaria immunity. Additionally, EMRA CD8+ T cells and CD56+CD8+ T cell clusters were associated with protection. In a cohort from a malaria-endemic area in Gabon, these CD8+ T cell clusters were also associated with parasitemia control in individuals with lifelong exposure to malaria. Upon stimulation with P. falciparum-infected erythrocytes, CD4+, γδ, and EMRA CD8+ T cells produced IFN-γ and/or TNF, indicating their ability to mediate responses that eliminate malaria parasites.


Sujet(s)
Lymphocytes T CD4+ , Lymphocytes T CD8+ , Vaccins contre le paludisme , Paludisme à Plasmodium falciparum , Plasmodium falciparum , Sporozoïtes , Adolescent , Adulte , Femelle , Humains , Mâle , Jeune adulte , Antipaludiques/usage thérapeutique , Antipaludiques/administration et posologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Chloroquine/usage thérapeutique , Chloroquine/pharmacologie , Europe , Européens , Gabon , Vaccins contre le paludisme/immunologie , Vaccins contre le paludisme/administration et posologie , Paludisme à Plasmodium falciparum/immunologie , Paludisme à Plasmodium falciparum/prévention et contrôle , Parasitémie/immunologie , Plasmodium falciparum/immunologie , Sporozoïtes/immunologie , Vaccination/méthodes , Vaccins atténués/immunologie , Vaccins atténués/administration et posologie , Peuples d'Afrique centrale
9.
ACS Infect Dis ; 10(6): 1904-1913, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38752809

RÉSUMÉ

Malaria parasites have a complex life cycle and undergo replication and population expansion within vertebrate hosts and mosquito vectors. These developmental transitions rely on changes in gene expression and chromatin reorganization that result in the activation and silencing of stage-specific genes. The ApiAp2 family of DNA-binding proteins plays an important role in regulating gene expression in malaria parasites. Here, we characterized the ApiAp2 protein in Plasmodium berghei, which we termed Ap2-D. In silico analysis revealed that Ap2-D has three beta-sheets followed by a helix at the C-terminus for DNA binding. Using gene tagging with 3XHA-mCherry, we found that Ap2-D is expressed in Plasmodium blood stages and is present in the parasite cytoplasm and nucleus. Surprisingly, our gene deletion study revealed a completely dispensable role for Ap2-D in the entirety of the P. berghei life cycle. Ap2-D KO parasites were found to grow in the blood successfully and progress through the mosquito midgut and salivary glands. Sporozoites isolated from mosquito salivary glands were infective for hepatocytes and achieved similar patency as WT in mice. We emphasize the importance of genetic validation of antimalarial drug targets before progressing them to drug discovery.


Sujet(s)
Étapes du cycle de vie , Plasmodium berghei , Protéines de protozoaire , Plasmodium berghei/génétique , Plasmodium berghei/croissance et développement , Plasmodium berghei/métabolisme , Animaux , Souris , Protéines de protozoaire/génétique , Protéines de protozoaire/métabolisme , Paludisme/parasitologie , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Sporozoïtes/croissance et développement , Sporozoïtes/métabolisme , Sporozoïtes/physiologie , Glandes salivaires/parasitologie , Vecteurs moustiques/parasitologie , Femelle , Anopheles/parasitologie , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Hépatocytes/parasitologie
10.
Parasit Vectors ; 17(1): 236, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38783366

RÉSUMÉ

BACKGROUND: Like other oviparous organisms, the gonotrophic cycle of mosquitoes is not complete until they have selected a suitable habitat to oviposit. In addition to the evolutionary constraints associated with selective oviposition behavior, the physiological demands relative to an organism's oviposition status also influence their nutrient requirement from the environment. Yet, studies that measure transmission potential (vectorial capacity or competence) of mosquito-borne parasites rarely consider whether the rates of parasite replication and development could be influenced by these constraints resulting from whether mosquitoes have completed their gonotrophic cycle. METHODS: Anopheles stephensi mosquitoes were infected with Plasmodium berghei, the rodent analog of human malaria, and maintained on 1% or 10% dextrose and either provided oviposition sites ('oviposited' herein) to complete their gonotrophic cycle or forced to retain eggs ('non-oviposited'). Transmission potential in the four groups was measured up to 27 days post-infection as the rates of (i) sporozoite appearance in the salivary glands ('extrinsic incubation period' or EIP), (ii) vector survival and (iii) sporozoite densities. RESULTS: In the two groups of oviposited mosquitoes, rates of sporozoite appearance and densities in the salivary glands were clearly dependent on sugar availability, with shorter EIP and higher sporozoite densities in mosquitoes fed 10% dextrose. In contrast, rates of appearance and densities in the salivary glands were independent of sugar concentrations in non-oviposited mosquitoes, although both measures were slightly lower than in oviposited mosquitoes fed 10% dextrose. Vector survival was higher in non-oviposited mosquitoes. CONCLUSIONS: Costs to parasite fitness and vector survival were buffered against changes in nutritional availability from the environment in non-oviposited but not oviposited mosquitoes. Taken together, these results suggest vectorial capacity for malaria parasites may be dependent on nutrient availability and oviposition/gonotrophic status and, as such, argue for more careful consideration of this interaction when estimating transmission potential. More broadly, the complex patterns resulting from physiological (nutrition) and evolutionary (egg-retention) trade-offs described here, combined with the ubiquity of selective oviposition behavior, implies the fitness of vector-borne pathogens could be shaped by selection for these traits, with implications for disease transmission and management. For instance, while reducing availability of oviposition sites and environmental sources of nutrition are key components of integrated vector management strategies, their abundance and distribution are under strong selection pressure from the patterns associated with climate change.


Sujet(s)
Anopheles , Paludisme , Vecteurs moustiques , Oviposition , Plasmodium berghei , Animaux , Anopheles/physiologie , Anopheles/parasitologie , Vecteurs moustiques/physiologie , Vecteurs moustiques/parasitologie , Femelle , Paludisme/transmission , Paludisme/parasitologie , Plasmodium berghei/physiologie , Glandes salivaires/parasitologie , Sporozoïtes/physiologie , Sucres/métabolisme , Souris
11.
Malar J ; 23(1): 151, 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38755636

RÉSUMÉ

BACKGROUND: Sporozoite invasion of hepatocytes is an essential step in the Plasmodium life-cycle and has similarities, at the cellular level, to merozoite invasion of erythrocytes. In the case of the Plasmodium blood-stage, efforts to identify host-pathogen protein-protein interactions have yielded important insights including vaccine candidates. In the case of sporozoite-hepatocyte invasion, the host-pathogen protein-protein interactions involved are poorly understood. METHODS: To gain a better understanding of the protein-protein interaction between the sporozoite ligands and host receptors, a systematic screen was performed. The previous Plasmodium falciparum and human surface protein ectodomain libraries were substantially extended, resulting in the creation of new libraries comprising 88 P. falciparum sporozoite protein coding sequences and 182 sequences encoding human hepatocyte surface proteins. Having expressed recombinant proteins from these sequences, a plate-based assay was used, capable of detecting low affinity interactions between recombinant proteins, modified for enhanced throughput, to screen the proteins for interactions. The novel interactions identified in the screen were characterized biochemically, and their essential role in parasite invasion was further elucidated using antibodies and genetically manipulated Plasmodium parasites. RESULTS: A total of 7540 sporozoite-hepatocyte protein pairs were tested under conditions capable of detecting interactions of at least 1.2 µM KD. An interaction between the human fibroblast growth factor receptor 4 (FGFR4) and the P. falciparum protein Pf34 is identified and reported here, characterizing its affinity and demonstrating the blockade of the interaction by reagents, including a monoclonal antibody. Furthermore, further interactions between Pf34 and a second P. falciparum rhoptry neck protein, PfRON6, and between human low-density lipoprotein receptor (LDLR) and the P. falciparum protein PIESP15 are identified. Conditional genetic deletion confirmed the essentiality of PfRON6 in the blood-stage, consistent with the important role of this protein in parasite lifecycle. Pf34 was refractory to attempted genetic modification. Antibodies to Pf34 abrogated the interaction and had a modest effect upon sporozoite invasion into primary human hepatocytes. CONCLUSION: Pf34 and PfRON6 may be members of a functionally important invasion complex which could be a target for future interventions. The modified interaction screening assay, protein expression libraries and P. falciparum mutant parasites reported here may be a useful tool for protein interaction discovery and antigen candidate screening which could be of wider value to the scientific community.


Sujet(s)
Hépatocytes , Plasmodium falciparum , Protéines de protozoaire , Sporozoïtes , Plasmodium falciparum/génétique , Plasmodium falciparum/métabolisme , Hépatocytes/parasitologie , Humains , Sporozoïtes/métabolisme , Protéines de protozoaire/génétique , Protéines de protozoaire/métabolisme , Interactions hôte-pathogène , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Récepteurs de surface cellulaire/génétique , Récepteurs de surface cellulaire/métabolisme , Interactions hôte-parasite , Liaison aux protéines
12.
Sci Rep ; 14(1): 12100, 2024 05 27.
Article de Anglais | MEDLINE | ID: mdl-38802488

RÉSUMÉ

Field-derived metrics are critical for effective control of malaria, particularly in sub-Saharan Africa where the disease kills over half a million people yearly. One key metric is entomological inoculation rate, a direct measure of transmission intensities, computed as a product of human biting rates and prevalence of Plasmodium sporozoites in mosquitoes. Unfortunately, current methods for identifying infectious mosquitoes are laborious, time-consuming, and may require expensive reagents that are not always readily available. Here, we demonstrate the first field-application of mid-infrared spectroscopy and machine learning (MIRS-ML) to swiftly and accurately detect Plasmodium falciparum sporozoites in wild-caught Anopheles funestus, a major Afro-tropical malaria vector, without requiring any laboratory reagents. We collected 7178 female An. funestus from rural Tanzanian households using CDC-light traps, then desiccated and scanned their heads and thoraces using an FT-IR spectrometer. The sporozoite infections were confirmed using enzyme-linked immunosorbent assay (ELISA) and polymerase chain reaction (PCR), to establish references for training supervised algorithms. The XGBoost model was used to detect sporozoite-infectious specimen, accurately predicting ELISA and PCR outcomes with 92% and 93% accuracies respectively. These findings suggest that MIRS-ML can rapidly detect P. falciparum in field-collected mosquitoes, with potential for enhancing surveillance in malaria-endemic regions. The technique is both fast, scanning 60-100 mosquitoes per hour, and cost-efficient, requiring no biochemical reactions and therefore no reagents. Given its previously proven capability in monitoring key entomological indicators like mosquito age, human blood index, and identities of vector species, we conclude that MIRS-ML could constitute a low-cost multi-functional toolkit for monitoring malaria risk and evaluating interventions.


Sujet(s)
Anopheles , Apprentissage machine , Paludisme à Plasmodium falciparum , Vecteurs moustiques , Plasmodium falciparum , Animaux , Anopheles/parasitologie , Paludisme à Plasmodium falciparum/épidémiologie , Paludisme à Plasmodium falciparum/diagnostic , Paludisme à Plasmodium falciparum/parasitologie , Plasmodium falciparum/isolement et purification , Vecteurs moustiques/parasitologie , Femelle , Humains , Tanzanie/épidémiologie , Sporozoïtes , Spectrophotométrie IR/méthodes , Spectroscopie infrarouge à transformée de Fourier/méthodes
13.
JCI Insight ; 9(11)2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38687615

RÉSUMÉ

A systems analysis was conducted to determine the potential molecular mechanisms underlying differential immunogenicity and protective efficacy results of a clinical trial of the radiation-attenuated whole-sporozoite PfSPZ vaccine in African infants. Innate immune activation and myeloid signatures at prevaccination baseline correlated with protection from P. falciparum parasitemia in placebo controls. These same signatures were associated with susceptibility to parasitemia among infants who received the highest and most protective PfSPZ vaccine dose. Machine learning identified spliceosome, proteosome, and resting DC signatures as prevaccination features predictive of protection after highest-dose PfSPZ vaccination, whereas baseline circumsporozoite protein-specific (CSP-specific) IgG predicted nonprotection. Prevaccination innate inflammatory and myeloid signatures were associated with higher sporozoite-specific IgG Ab response but undetectable PfSPZ-specific CD8+ T cell responses after vaccination. Consistent with these human data, innate stimulation in vivo conferred protection against infection by sporozoite injection in malaria-naive mice while diminishing the CD8+ T cell response to radiation-attenuated sporozoites. These data suggest a dichotomous role of innate stimulation for malaria protection and induction of protective immunity by whole-sporozoite malaria vaccines. The uncoupling of vaccine-induced protective immunity achieved by Abs from more protective CD8+ T cell responses suggests that PfSPZ vaccine efficacy in malaria-endemic settings may be constrained by opposing antigen presentation pathways.


Sujet(s)
Immunité innée , Vaccins contre le paludisme , Paludisme à Plasmodium falciparum , Plasmodium falciparum , Sporozoïtes , Vaccins atténués , Vaccins contre le paludisme/immunologie , Vaccins contre le paludisme/administration et posologie , Immunité innée/immunologie , Humains , Animaux , Paludisme à Plasmodium falciparum/prévention et contrôle , Paludisme à Plasmodium falciparum/immunologie , Plasmodium falciparum/immunologie , Souris , Vaccins atténués/immunologie , Vaccins atténués/administration et posologie , Sporozoïtes/immunologie , Sporozoïtes/effets des radiations , Lymphocytes T CD8+/immunologie , Nourrisson , Protéines de protozoaire/immunologie , Anticorps antiprotozoaires/immunologie , Femelle , Parasitémie/immunologie , Parasitémie/prévention et contrôle , Immunoglobuline G/immunologie , Immunoglobuline G/sang ,
14.
Microbes Infect ; 26(5-6): 105343, 2024.
Article de Anglais | MEDLINE | ID: mdl-38670216

RÉSUMÉ

Hemozoin is a crystal synthesized by Plasmodium parasites during hemoglobin digestion in the erythrocytic stage. The hemozoin released when the parasites egress from the red blood cell, which is complexed with parasite DNA, is cleared from the circulation by circulating and tissue-resident monocytes and macrophages, respectively. Recently, we reported that intravenous administration of purified hemozoin complexed with Plasmodium berghei DNA (HzPbDNA) resulted in an innate immune response that blocked liver stage development of sporozoites that was dose-dependent and time-limited. Here, we further characterize the organismal, cellular, and molecular events associated with this protective innate response in the liver and report that a large proportion of the IV administered HzPbDNA localized to F4/80+ cells in the liver and that the rapid and strong protection against liver-stage development waned quickly such that by 1 week post-HzPbDNA treatment animals were fully susceptible to infection. RNAseq of the liver after IV administration of HzPbDNA demonstrated that the rapid and robust induction of genes associated with the acute phase response, innate immune activation, cellular recruitment, and IFN-γ signaling observed at day 1 was largely absent at day 7. RNAseq analysis implicated NK cells as the major cellular source of IFN-γ. In vivo cell depletion and IFN-γ neutralization experiments supported the hypothesis that tissue-resident macrophages and NK cells are major contributors to the protective response and the NK cell-derived IFN-γ is key to induction of the mechanisms that block sporozoite development in the liver. These findings advance our understanding of the innate immune responses that prevent liver stage malaria infection.


Sujet(s)
Hémoprotéines , Immunité innée , Interféron gamma , Foie , Paludisme , Plasmodium berghei , Sporozoïtes , Animaux , Plasmodium berghei/immunologie , Sporozoïtes/immunologie , Paludisme/immunologie , Paludisme/prévention et contrôle , Paludisme/parasitologie , Hémoprotéines/immunologie , Souris , Foie/parasitologie , Foie/immunologie , Interféron gamma/immunologie , Interféron gamma/métabolisme , Souris de lignée C57BL , Macrophages/immunologie , Macrophages/parasitologie , ADN des protozoaires/génétique , Femelle
15.
Front Cell Infect Microbiol ; 14: 1367359, 2024.
Article de Anglais | MEDLINE | ID: mdl-38660488

RÉSUMÉ

Cryptosporidium parvum is a common cause of a zoonotic disease and a main cause of diarrhea in newborns. Effective drugs or vaccines are still lacking. Oocyst is the infective form of the parasite; after its ingestion, the oocyst excysts and releases four sporozoites into the host intestine that rapidly attack the enterocytes. The membrane protein CpRom1 is a large rhomboid protease that is expressed by sporozoites and recognized as antigen by the host immune system. In this study, we observed the release of CpRom1 with extracellular vesicles (EVs) that was not previously described. To investigate this phenomenon, we isolated and resolved EVs from the excystation medium by differential ultracentrifugation. Fluorescence flow cytometry and transmission electron microscopy (TEM) experiments identified two types of sporozoite-derived vesicles: large extracellular vesicles (LEVs) and small extracellular vesicles (SEVs). Nanoparticle tracking analysis (NTA) revealed mode diameter of 181 nm for LEVs and 105 nm for SEVs, respectively. Immunodetection experiments proved the presence of CpRom1 and the Golgi protein CpGRASP in LEVs, while immune-electron microscopy trials demonstrated the localization of CpRom1 on the LEVs surface. TEM and scanning electron microscopy (SEM) showed that LEVs were generated by means of the budding of the outer membrane of sporozoites; conversely, the origin of SEVs remained uncertain. Distinct protein compositions were observed between LEVs and SEVs as evidenced by their corresponding electrophoretic profiles. Indeed, a dedicated proteomic analysis identified 5 and 16 proteins unique for LEVs and SEVs, respectively. Overall, 60 proteins were identified in the proteome of both types of vesicles and most of these proteins (48 in number) were already identified in the molecular cargo of extracellular vesicles from other organisms. Noteworthy, we identified 12 proteins unique to Cryptosporidium spp. and this last group included the immunodominant parasite antigen glycoprotein GP60, which is one of the most abundant proteins in both LEVs and SEVs.


Sujet(s)
Cryptosporidium parvum , Vésicules extracellulaires , Protéines de protozoaire , Sporozoïtes , Vésicules extracellulaires/métabolisme , Cryptosporidium parvum/métabolisme , Sporozoïtes/métabolisme , Protéines de protozoaire/métabolisme , Protéines de protozoaire/analyse , Microscopie électronique à transmission , Animaux , Cryptosporidiose/parasitologie , Humains , Protéome/analyse , Protéomique , Cytométrie en flux
16.
Malar J ; 23(1): 106, 2024 Apr 17.
Article de Anglais | MEDLINE | ID: mdl-38632607

RÉSUMÉ

BACKGROUND: To gain a deeper understanding of protective immunity against relapsing malaria, this study examined sporozoite-specific T cell responses induced by a chemoprophylaxis with sporozoite (CPS) immunization in a relapsing Plasmodium cynomolgi rhesus macaque model. METHODS: The animals received three CPS immunizations with P. cynomolgi sporozoites, administered by mosquito bite, while under two anti-malarial drug regimens. Group 1 (n = 6) received artesunate/chloroquine (AS/CQ) followed by a radical cure with CQ plus primaquine (PQ). Group 2 (n = 6) received atovaquone-proguanil (AP) followed by PQ. After the final immunization, the animals were challenged with intravenous injection of 104 P. cynomolgi sporozoites, the dose that induced reliable infection and relapse rate. These animals, along with control animals (n = 6), were monitored for primary infection and subsequent relapses. Immunogenicity blood draws were done after each of the three CPS session, before and after the challenge, with liver, spleen and bone marrow sampling and analysis done after the challenge. RESULTS: Group 2 animals demonstrated superior protection, with two achieving protection and two experiencing partial protection, while only one animal in group 1 had partial protection. These animals displayed high sporozoite-specific IFN-γ T cell responses in the liver, spleen, and bone marrow after the challenge with one protected animal having the highest frequency of IFN-γ+ CD8+, IFN-γ+ CD4+, and IFN-γ+ γδ T cells in the liver. Partially protected animals also demonstrated a relatively high frequency of IFN-γ+ CD8+, IFN-γ+ CD4+, and IFN-γ+ γδ T cells in the liver. It is important to highlight that the second animal in group 2, which experienced protection, exhibited deficient sporozoite-specific T cell responses in the liver while displaying average to high T cell responses in the spleen and bone marrow. CONCLUSIONS: This research supports the notion that local liver T cell immunity plays a crucial role in defending against liver-stage infection. Nevertheless, there is an instance where protection occurs independently of T cell responses in the liver, suggesting the involvement of the liver's innate immunity. The relapsing P. cynomolgi rhesus macaque model holds promise for informing the development of vaccines against relapsing P. vivax.


Sujet(s)
Atovaquone , Vaccins contre le paludisme , Plasmodium cynomolgi , Proguanil , Animaux , Primaquine/usage thérapeutique , Sporozoïtes , Macaca mulatta , Immunisation , Chimioprévention , Lymphocytes T CD8+ , Association médicamenteuse
17.
Malar J ; 23(1): 111, 2024 Apr 19.
Article de Anglais | MEDLINE | ID: mdl-38641838

RÉSUMÉ

BACKGROUND: Sporozoites (SPZ), the infective form of Plasmodium falciparum malaria, can be inoculated into the human host skin by Anopheline mosquitoes. These SPZ migrate at approximately 1 µm/s to find a blood vessel and travel to the liver where they infect hepatocytes and multiply. In the skin they are still low in number (50-100 SPZ) and vulnerable to immune attack by antibodies and skin macrophages. This is why whole SPZ and SPZ proteins are used as the basis for most malaria vaccines currently deployed and undergoing late clinical testing. Mosquitoes typically inoculate SPZ into a human host between 14 and 25 days after their previous infective blood meal. However, it is unknown whether residing time within the mosquito affects SPZ condition, infectivity or immunogenicity. This study aimed to unravel how the age of P. falciparum SPZ in salivary glands (14, 17, or 20 days post blood meal) affects their infectivity and the ensuing immune responses. METHODS: SPZ numbers, viability by live/dead staining, motility using dedicated sporozoite motility orienting and organizing tool software (SMOOT), and infectivity of HC-04.j7 liver cells at 14, 17 and 20 days after mosquito feeding have been investigated. In vitro co-culture assays with SPZ stimulated monocyte-derived macrophages (MoMɸ) and CD8+ T-cells, analysed by flow cytometry, were used to investigate immune responses. RESULTS: SPZ age did not result in different SPZ numbers or viability. However, a markedly different motility pattern, whereby motility decreased from 89% at day 14 to 80% at day 17 and 71% at day 20 was observed (p ≤ 0.0001). Similarly, infectivity of day 20 SPZ dropped to ~ 50% compared with day 14 SPZ (p = 0.004). MoMɸ were better able to take up day 14 SPZ than day 20 SPZ (from 7.6% to 4.1%, p = 0.03) and displayed an increased expression of pro-inflammatory CD80, IL-6 (p = 0.005), regulatory markers PDL1 (p = 0.02), IL-10 (p = 0.009) and cytokines upon phagocytosis of younger SPZ. Interestingly, co-culture of these cells with CD8+ T-cells revealed a decreased expression of activation marker CD137 and cytokine IFNγ compared to their day 20 counterparts. These findings suggest that older (day 17-20) P. falciparum SPZ are less infectious and have decreased immune regulatory potential. CONCLUSION: Overall, this data is a first step in enhancing the understanding of how mosquito residing time affects P. falciparum SPZ and could impact the understanding of the P. falciparum infectious reservoir and the potency of whole SPZ vaccines.


Sujet(s)
Culicidae , Vaccins contre le paludisme , Paludisme à Plasmodium falciparum , Animaux , Humains , Sporozoïtes , Lymphocytes T CD8+ , Vieillissement , Plasmodium falciparum
18.
Am J Trop Med Hyg ; 110(5): 892-901, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38531102

RÉSUMÉ

Malaria eradication efforts prioritize safe and efficient vaccination strategies, although none with high-level efficacy against malaria infection are yet available. Among several vaccine candidates, Sanaria® PfSPZ Vaccine and Sanaria PfSPZ-CVac are, respectively, live radiation- and chemo-attenuated sporozoite vaccines designed to prevent infection with Plasmodium falciparum, the leading cause of malaria-related morbidity and mortality. We are conducting a randomized normal saline placebo-controlled trial called IDSPZV1 that will analyze the safety, tolerability, immunogenicity, and efficacy of PfSPZ Vaccine and PfSPZ-CVac administered pre-deployment to malaria-naive Indonesian soldiers assigned to temporary duties in a high malaria transmission area. We describe the manifold challenges of enrolling and immunizing 345 soldier participants at their home base in western Indonesia before their nearly 6,000-km voyage to eastern Indonesia, where they are being monitored for incident P. falciparum and Plasmodium vivax malaria cases during 9 months of exposure. The unique regulatory, ethical, and operational complexities of this trial demonstrate the importance of thorough planning, frequent communication, and close follow-up with stakeholders. Effective engagement with the military community and the ability to adapt to unanticipated events have proven key to the success of this trial.


Sujet(s)
Vaccins contre le paludisme , Paludisme à Plasmodium falciparum , Paludisme à Plasmodium vivax , Personnel militaire , Plasmodium falciparum , Sporozoïtes , Vaccins atténués , Humains , Vaccins contre le paludisme/immunologie , Vaccins contre le paludisme/usage thérapeutique , Vaccins contre le paludisme/administration et posologie , Indonésie/épidémiologie , Paludisme à Plasmodium falciparum/prévention et contrôle , Paludisme à Plasmodium falciparum/épidémiologie , Sporozoïtes/immunologie , Vaccins atténués/immunologie , Vaccins atténués/usage thérapeutique , Plasmodium falciparum/immunologie , Paludisme à Plasmodium vivax/prévention et contrôle , Paludisme à Plasmodium vivax/épidémiologie , Mâle , Adulte , Jeune adulte , Plasmodium vivax/immunologie , Femelle
19.
Malar J ; 23(1): 81, 2024 Mar 16.
Article de Anglais | MEDLINE | ID: mdl-38493098

RÉSUMÉ

BACKGROUND: Vector surveillance is among the World Health Organization global vector control response (2017-2030) pillars. Human landing catches are a gold standard but difficult to implement and potentially expose collectors to malaria infection. Other methods like light traps, pyrethrum spray catches and aspiration are less expensive and less risky to collectors. METHODS: Three mosquito sampling methods (UV light traps, CDC light traps and Prokopack aspiration) were evaluated against human landing catches (HLC) in two villages of Rarieda sub-county, Siaya County, Kenya. UV-LTs, CDC-LTs and HLCs were conducted hourly between 17:00 and 07:00. Aspiration was done indoors and outdoors between 07:00 and 11:00 a.m. Analyses of mosquito densities, species abundance and sporozoite infectivity were performed across all sampling methods. Species identification PCR and ELISAs were done for Anopheles gambiae and Anopheles funestus complexes and data analysis was done in R. RESULTS: Anopheles mosquitoes sampled from 608 trapping efforts were 5,370 constituting 70.3% Anopheles funestus sensu lato (s.l.), 19.7% Anopheles coustani and 7.2% An. gambiae s.l. 93.8% of An. funestus s.l. were An. funestus sensu stricto (s.s.) and 97.8% of An. gambiae s.l. were Anopheles arabiensis. Only An. funestus were sporozoite positive with 3.1% infection prevalence. Indoors, aspiration captured higher An. funestus (mean = 6.74; RR = 8.83, P < 0.001) then UV-LT (mean = 3.70; RR = 3.97, P < 0.001) and CDC-LT (mean = 1.74; RR = 1.89, P = 0.03) compared to HLC. UV-LT and CDC-LT indoors captured averagely 0.18 An. arabiensis RR = 5.75, P = 0.028 and RR = 5.87, P = 0.028 respectively. Outdoors, UV-LT collected significantly higher Anopheles mosquitoes compared to HLC (An. funestus: RR = 5.18, P < 0.001; An. arabiensis: RR = 15.64, P = 0.009; An. coustani: RR = 11.65, P < 0.001). Anopheles funestus hourly biting indoors in UV-LT and CDC-LT indicated different peaks compared to HLC. CONCLUSIONS: Anopheles funestus remains the predominant mosquito species. More mosquitoes were collected using aspiration, CDC-LTs and UV-LTs indoors and UV-LTs and CD-LTs outdoors compared to HLCs. UV-LTs collected more mosquitoes than CDC-LTs. The varied trends observed at different times of the night suggest that these methods collect mosquitoes with diverse activities and care must be taken when interpreting the results.


Sujet(s)
Anopheles , Paludisme , Animaux , Humains , Anopheles/physiologie , Kenya/épidémiologie , Vecteurs moustiques/physiologie , Comportement alimentaire , Sporozoïtes , Lutte contre les moustiques/méthodes
20.
Malar J ; 23(1): 72, 2024 Mar 11.
Article de Anglais | MEDLINE | ID: mdl-38468292

RÉSUMÉ

BACKGROUND: Recently, bacterial endosymbiont, including Wolbachia and Microsporidia were found to limit the infection of Anopheles mosquitoes with Plasmodium falciparum. This study aimed to investigate the natural presence of key transmission-blocking endosymbionts in Anopheles gambiae and Anopheles coluzzii in Southern Benin. METHODS: The present study was conducted in seven communes (Cotonou, Porto-Novo, Aguégués, Ifangni, Pobè Athiémé, and Grand-Popo) of Southern Benin. Anopheles were collected using indoor/outdoor Human Landing Catches (HLCs) and Pyrethrum Spray Catches (PSCs). Following morphological identification, PCR was used to identify An. gambiae sensu lato (s.l.) to species level and to screen for the presence of both Wolbachia and Microsporidia. Plasmodium falciparum sporozoite infection was also assessed using ELISA. RESULTS: Overall, species composition in An. gambiae s.l. was 53.7% An. coluzzii, while the remainder was An. gambiae sensu stricto (s.s.). Combined data of the two sampling techniques revealed a mean infection prevalence with Wolbachia of 5.1% (95% CI 0.90-18.6) and 1.3% (95% CI 0.07-7.8) in An. gambiae s.s. and An. coluzzii, respectively. The mean infection prevalence with Microsporidia was 41.0% (95% CI 25.9-57.8) for An. gambiae s.s. and 57.0% (95% CI 45.4-67.9) for An. coluzzii. Wolbachia was only observed in Ifangni, Pobè, and Cotonou, while Microsporidia was detected in all study communes. Aggregated data for HLCs and PSCs showed a sporozoite rate (SR) of 0.80% (95% CI 0.09-2.87) and 0.69% (95% CI 0.09-2.87) for An. gambiae and An. coluzzii, respectively, with a mean of 0.74% (95% CI 0.20-1.90). Of the four individual mosquitoes which harboured P. falciparum, none were also infected with Wolbachia and one contained Microsporidia. CONCLUSIONS: The present study is the first report of natural infections of field-collected An. gambiae s.l. populations from Benin with Wolbachia and Microsporidia. Sustained efforts should be made to widen the spectrum of bacteria identified in mosquitoes, with the potential to develop endosymbiont-based control tools; such interventions could be the game-changer in the control of malaria and arboviral disease transmission.


Sujet(s)
Anopheles , Paludisme à Plasmodium falciparum , Pyréthrines , Wolbachia , Animaux , Humains , Bénin/épidémiologie , Études transversales , Vecteurs moustiques , Paludisme à Plasmodium falciparum/épidémiologie , Sporozoïtes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE