Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 511
Filter
1.
Front Cell Infect Microbiol ; 14: 1382228, 2024.
Article in English | MEDLINE | ID: mdl-38698904

ABSTRACT

Background: Tick-borne pathogen (TBP) surveillance studies often use whole-tick homogenates when inferring tick-pathogen associations. However, localized TBP infections within tick tissues (saliva, hemolymph, salivary glands, and midgut) can inform pathogen transmission mechanisms and are key to disentangling pathogen detection from vector competence. Methods: We screened 278 camel blood samples and 504 tick tissue samples derived from 126 camel ticks sampled in two Kenyan counties (Laikipia and Marsabit) for Anaplasma, Ehrlichia, Coxiella, Rickettsia, Theileria, and Babesia by PCR-HRM analysis. Results: Candidatus Anaplasma camelii infections were common in camels (91%), but absent in all samples from Rhipicephalus pulchellus, Amblyomma gemma, Hyalomma dromedarii, and Hyalomma rufipes ticks. We detected Ehrlichia ruminantium in all tissues of the four tick species, but Rickettsia aeschlimannii was only found in Hy. rufipes (all tissues). Rickettsia africae was highest in Am. gemma (62.5%), mainly in the hemolymph (45%) and less frequently in the midgut (27.5%) and lowest in Rh. pulchellus (29.4%), where midgut and hemolymph detection rates were 17.6% and 11.8%, respectively. Similarly, in Hy. dromedarii, R. africae was mainly detected in the midgut (41.7%) but was absent in the hemolymph. Rickettsia africae was not detected in Hy. rufipes. No Coxiella, Theileria, or Babesia spp. were detected in this study. Conclusions: The tissue-specific localization of R. africae, found mainly in the hemolymph of Am. gemma, is congruent with the role of this tick species as its transmission vector. Thus, occurrence of TBPs in the hemolymph could serve as a predictor of vector competence of TBP transmission, especially in comparison to detection rates in the midgut, from which they must cross tissue barriers to effectively replicate and disseminate across tick tissues. Further studies should focus on exploring the distribution of TBPs within tick tissues to enhance knowledge of TBP epidemiology and to distinguish competent vectors from dead-end hosts.


Subject(s)
Babesia , Camelus , Ehrlichia , Theileria , Ticks , Animals , Kenya/epidemiology , Camelus/parasitology , Camelus/microbiology , Theileria/isolation & purification , Theileria/genetics , Babesia/isolation & purification , Babesia/genetics , Ehrlichia/isolation & purification , Ehrlichia/genetics , Ticks/microbiology , Ticks/parasitology , Tick-Borne Diseases/microbiology , Tick-Borne Diseases/epidemiology , Tick-Borne Diseases/parasitology , Anaplasma/isolation & purification , Anaplasma/genetics , Rickettsia/isolation & purification , Rickettsia/genetics , Coxiella/isolation & purification , Coxiella/genetics , Hemolymph/microbiology , Hemolymph/parasitology , Salivary Glands/microbiology , Salivary Glands/parasitology
2.
Parasit Vectors ; 17(1): 236, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783366

ABSTRACT

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.


Subject(s)
Anopheles , Malaria , Mosquito Vectors , Oviposition , Plasmodium berghei , Animals , Anopheles/physiology , Anopheles/parasitology , Mosquito Vectors/physiology , Mosquito Vectors/parasitology , Female , Malaria/transmission , Malaria/parasitology , Plasmodium berghei/physiology , Salivary Glands/parasitology , Sporozoites/physiology , Sugars/metabolism , Mice
3.
Parasitol Res ; 122(7): 1689-1693, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37099049

ABSTRACT

The competence of insect vectors to transmit diseases plays a key role in host-parasite interactions and in the dynamics of avian malaria and other haemosporidian infections (Apicomplexa, Haemosporida). However, the presence of parasite DNA in the body of blood-sucking insects does not always constitute evidence for their competence as vectors. In this study, we investigate the susceptibility of wild-caught mosquitoes (Culex spp.) to complete sporogony of Plasmodium relictum (cyt b lineage SGS1) isolated from great tits (Parus major L., 1758). Adult female mosquitoes were collected with a CO2 bait trap overnight. A set of 50 mosquitoes was allowed to feed for 3 h at night on a single great tit infected with P. relictum. This trial was repeated on 6 different birds. The bloodfed mosquitoes that survived (n = 68) were dissected within 1-2 days (for ookinetes, n = 10) and 10-33 days post infection (for oocysts and sporozoites, n = 58) in order to confirm the respective parasite stages in their organs. The experiment confirmed the successful development of P. relictum (cyt b lineage SGS1) to the stage of sporozoites in Culex pipiens L., 1758 (n = 27) and in Culex modestus (n = 2). Our study provides the first evidence that C. modestus is a competent vector of P. relictum isolated from great tits, suggesting that this mosquito species could also play a role in the natural transmission of avian malaria.


Subject(s)
Culex , Culicidae , Malaria, Avian , Passeriformes , Plasmodium , Animals , Female , Malaria, Avian/parasitology , Culex/parasitology , Sporozoites , Cytochromes b/genetics , Mosquito Vectors/parasitology , Plasmodium/genetics , Salivary Glands/parasitology , Passeriformes/parasitology
4.
PLoS Pathog ; 19(3): e1011269, 2023 03.
Article in English | MEDLINE | ID: mdl-36996244

ABSTRACT

Trypanosoma brucei spp. develop into mammalian-infectious metacyclic trypomastigotes inside tsetse salivary glands. Besides acquiring a variant surface glycoprotein (VSG) coat, little is known about the metacyclic expression of invariant surface antigens. Proteomic analyses of saliva from T. brucei-infected tsetse flies identified, in addition to VSG and Brucei Alanine-Rich Protein (BARP) peptides, a family of glycosylphosphatidylinositol (GPI)-anchored surface proteins herein named as Metacyclic Invariant Surface Proteins (MISP) because of its predominant expression on the surface of metacyclic trypomastigotes. The MISP family is encoded by five paralog genes with >80% protein identity, which are exclusively expressed by salivary gland stages of the parasite and peak in metacyclic stage, as shown by confocal microscopy and immuno-high resolution scanning electron microscopy. Crystallographic analysis of a MISP isoform (MISP360) and a high confidence model of BARP revealed a triple helical bundle architecture commonly found in other trypanosome surface proteins. Molecular modelling combined with live fluorescent microscopy suggests that MISP N-termini are potentially extended above the metacyclic VSG coat, and thus could be tested as a transmission-blocking vaccine target. However, vaccination with recombinant MISP360 isoform did not protect mice against a T. brucei infectious tsetse bite. Lastly, both CRISPR-Cas9-driven knock out and RNAi knock down of all MISP paralogues suggest they are not essential for parasite development in the tsetse vector. We suggest MISP may be relevant during trypanosome transmission or establishment in the vertebrate's skin.


Subject(s)
Parasites , Trypanosoma brucei brucei , Trypanosoma , Animals , Mice , Trypanosoma brucei brucei/genetics , Membrane Proteins , Alanine , Proteomics , Salivary Glands/parasitology , Mammals , Membrane Glycoproteins
5.
PLoS Pathog ; 18(10): e1010881, 2022 10.
Article in English | MEDLINE | ID: mdl-36223382

ABSTRACT

Mosquito saliva is a vehicle for the transmission of vector borne pathogens such as Plasmodium parasites and different arboviruses. Despite the key role of the salivary glands in the process of disease transmission, knowledge of host-pathogen interactions taking place within this organ is very limited. To improve the experimental tractability of the salivary glands, we have generated fluorescent reporter lines in the African malaria mosquito Anopheles coluzzii using the salivary gland-specific promoters of the anopheline antiplatelet protein (AAPP), the triple functional domain protein (TRIO) and saglin (SAG) coding genes. Promoter activity was specifically observed in the distal-lateral lobes or in the median lobe of the salivary glands. Besides a comparison of the expression patterns of the selected promoters, the fluorescent probes allowed us to evaluate the inducibility of the selected promoters upon blood feeding and to measure intracellular redox changes. We also combined the aapp-DsRed fluorescent reporter line with a pigmentation-deficient yellow(-) mosquito mutant to assess the feasibility of in vivo microscopy of parasitized salivary glands. This combination allowed locating the salivary gland through the cuticle and imaging of individual sporozoites in vivo, which facilitates live imaging studies of salivary gland colonization by Plasmodium sporozoites.


Subject(s)
Anopheles , Malaria , Plasmodium , Animals , Anopheles/genetics , Anopheles/parasitology , Biology , Fluorescent Dyes , Malaria/parasitology , Mosquito Vectors/genetics , Mosquito Vectors/parasitology , Salivary Glands/parasitology , Sporozoites
6.
Malar J ; 21(1): 264, 2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36100902

ABSTRACT

BACKGROUND: Sporozoites isolated from the salivary glands of Plasmodium-infected mosquitoes are a prerequisite for several basic and pre-clinical applications. Although salivary glands are pooled to maximize sporozoite recovery, insufficient yields pose logistical and analytical hurdles; thus, predicting yields prior to isolation would be valuable. Preceding oocyst densities in the midgut is an obvious candidate. However, it is unclear whether current understanding of its relationship with sporozoite densities can be used to maximize yields, or whether it can capture the potential density-dependence in rates of sporozoite invasion of the salivary glands. METHODS: This study presents a retrospective analysis of Anopheles stephensi mosquitoes infected with two strains of the rodent-specific Plasmodium berghei. Mean oocyst densities were estimated in the midguts earlier in the infection (11-15 days post-blood meal), with sporozoites pooled from the salivary glands later in the infection (17-29 days). Generalized linear mixed effects models were used to determine if (1) mean oocyst densities can predict sporozoite yields from pooled salivary glands, (2) whether these densities can capture differences in rates of sporozoite invasion of salivary glands, and (3), if the interaction between oocyst densities and time could be leveraged to boost overall yields. RESULTS: The non-linear effect of mean oocyst densities confirmed the role of density-dependent constraints in limiting yields beyond certain oocyst densities. Irrespective of oocyst densities however, the continued invasion of salivary glands by the sporozoites boosted recoveries over time (17-29 days post-blood meal) for either parasite strain. CONCLUSIONS: Sporozoite invasion of the salivary glands over time can be leveraged to maximize yields for P. berghei. In general, however, invasion of the salivary glands over time is a critical fitness determinant for all Plasmodium species (extrinsic incubation period, EIP). Thus, delaying sporozoite collection could, in principle, substantially reduce dissection effort for any parasite within the genus, with the results also alluding to the potential for changes in sporozoites densities over time to modify infectivity for the next host.


Subject(s)
Anopheles , Sporozoites , Animals , Anopheles/parasitology , Plasmodium berghei , Retrospective Studies , Salivary Glands/parasitology
7.
Parasitol Int ; 87: 102515, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34781013

ABSTRACT

Black flies (Diptera: Simuliidae) are among the most bothersome blood-sucking dipterans causing severe irritation and distress to poultry, wild birds, animals, and humans globally. These insects are vectors of viruses, bacteria, parasitic protozoans, and nematodes of humans and animals. Parasitic protozoa belonging to Haemosporida (Apicomplexa) are distributed worldwide and black flies are the principal vectors of avian haemosporidian parasites of the genus Leucocytozoon, a common parasite of birds. Based on the detection of parasite DNA in insects, 13 black fly species were reported to be potential vectors of Leucocytozoon in Europe. Information about which species of Simulium can play a role in the transmission of Leucocytozoon parasites is insufficient and needs to be developed. The aim of our study was to determine which black fly species are involved in the transmission of Leucocytozoon parasites in the Eastern Europe. The black fly females were collected in Lithuania using entomological net. They were morphologically identified, dissected to prepare salivary glands preparations, and then screened for the presence of Leucocytozoon parasites using microscopy and PCR-based methods. In all, we collected 437 black fly females belonging to eight species. The DNA of Leucocytozoon (genetic lineage lCOCO18) was detected in one of analysed females identified as Simulium maculatum. All salivary gland preparations were negative for the presence of Leucocytozoon sporozoites. Our results included S. maculatum as a potential vector of Leucocytozoon parasites. Increasing the knowledge on vector ecology, behaviour and improving collection methods may be the key to understand the evolution and diversity of these parasites.


Subject(s)
Bird Diseases/parasitology , Haemosporida/physiology , Insect Vectors/parasitology , Protozoan Infections, Animal/transmission , Simuliidae/parasitology , Animals , Bird Diseases/transmission , Birds , DNA/isolation & purification , DNA, Protozoan/isolation & purification , Female , Haemosporida/genetics , Humans , Lithuania , Phylogeny , Protozoan Infections, Animal/parasitology , Salivary Glands/parasitology
8.
Microbiol Spectr ; 9(2): e0085221, 2021 10 31.
Article in English | MEDLINE | ID: mdl-34668767

ABSTRACT

The emergence of resistance to antimalarials has prompted the steady switch to novel therapies for decades. Withdrawal of antimalarials, such as chloroquine in sub-Saharan Africa in the late 1990s, led to rapid declines in the prevalence of resistance markers after a few years, raising the possibility of reintroducing them for malaria treatment. Here, we provide evidence that the mosquito vector plays a crucial role in maintaining parasite genetic diversity. We followed the transmission dynamics of Plasmodium falciparum parasites through its vector in natural infections from gametocytes contained in the blood of asymptomatic volunteers until sporozoites subsequently developed in the mosquito salivary glands. We did not find any selection of the mutant or wild-type pfcrt 76 allele during development in the Anopheles mosquito vector. However, microsatellite genotyping indicated that minority genotypes were favored during transmission through the mosquito. The analysis of changes in the proportions of mutant and wild-type pfcrt 76 alleles showed that, regardless of the genotype, the less-represented allele in the gametocyte population was more abundant in mosquito salivary glands, indicating a selective advantage of the minority allele in the vector. Selection of minority genotypes in the vector would explain the persistence of drug-resistant alleles in the absence of drug pressure in areas with high malaria endemicity and high genetic diversity. Our results may have important epidemiological implications, as they predict the rapid re-emergence and spread of resistant genotypes if antimalarials that had previously selected resistant parasites are reintroduced for malaria prevention or treatment. IMPORTANCE Drug selection pressure in malaria patients is the cause of the emergence of resistant parasites. Resistance imposes a fitness cost for parasites in untreated infections, so withdrawal of the drug leads to the return of susceptible parasites. Little is known about the role of the malaria vector in this phenomenon. In an experimental study conducted in Cameroon, an area of high malaria transmission, we showed that the vector did not favor the parasites based on sensitivity or resistance criteria, but it did favor the selection of minority clones. This finding shows that the vector increases the diversity of plasmodial populations and could play an important role in falciparum malaria epidemiology by maintaining resistant clones despite the absence of therapeutic pressure.


Subject(s)
Anopheles/parasitology , Drug Resistance/genetics , Malaria, Falciparum/epidemiology , Malaria, Falciparum/transmission , Mosquito Vectors/parasitology , Plasmodium falciparum/drug effects , Alleles , Animals , Antimalarials/therapeutic use , Cameroon/epidemiology , Chloroquine/therapeutic use , Genetic Variation/genetics , Genotype , Malaria, Falciparum/drug therapy , Membrane Transport Proteins/genetics , Microsatellite Repeats/genetics , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Salivary Glands/parasitology , Selection, Genetic/genetics
9.
PLoS Negl Trop Dis ; 15(9): e0009504, 2021 09.
Article in English | MEDLINE | ID: mdl-34543277

ABSTRACT

Trypanosoma brucei ssp., unicellular parasites causing human and animal trypanosomiasis, are transmitted between mammals by tsetse flies. Periodic changes in variant surface glycoproteins (VSG), which form the parasite coat in the mammal, allow them to evade the host immune response. Different isolates of T. brucei show heterogeneity in their repertoires of VSG genes and have single nucleotide polymorphisms and indels that can impact on genome editing. T. brucei brucei EATRO1125 (AnTaR1 serodeme) is an isolate that is used increasingly often because it is pleomorphic in mammals and fly transmissible, two characteristics that have been lost by the most commonly used laboratory stocks. We present a genome assembly of EATRO1125, including contigs for the intermediate chromosomes and minichromosomes that serve as repositories of VSG genes. In addition, de novo transcriptome assemblies were performed using Illumina sequences from tsetse-derived trypanosomes. Reads of 150 bases enabled closely related members of multigene families to be discriminated. This revealed that the transcriptome of midgut-derived parasites is dynamic, starting with the expression of high affinity hexose transporters and glycolytic enzymes and then switching to proline uptake and catabolism. These changes resemble the transition from early to late procyclic forms in culture. Further metabolic reprogramming, including upregulation of tricarboxylic acid cycle enzymes, occurs in the proventriculus. Many transcripts upregulated in the salivary glands encode surface proteins, among them 7 metacyclic VSGs, multiple BARPs and GCS1/HAP2, a marker for gametes. A novel family of transmembrane proteins, containing polythreonine stretches that are predicted to be O-glycosylation sites, was also identified. Finally, RNA-Seq data were used to create an optimised annotation file with 5' and 3' untranslated regions accurately mapped for 9302 genes. We anticipate that this will be of use in identifying transcripts obtained by single cell sequencing technologies.


Subject(s)
DNA, Protozoan/genetics , Gene Expression Regulation, Developmental/physiology , Insect Vectors/parasitology , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/physiology , Tsetse Flies/parasitology , Animals , Energy Metabolism , Gene Expression Profiling , Genome, Protozoan , Host-Parasite Interactions , Protozoan Proteins/genetics , RNA-Seq , Salivary Glands/parasitology
10.
PLoS Pathog ; 17(9): e1009904, 2021 09.
Article in English | MEDLINE | ID: mdl-34543350

ABSTRACT

The long and complex Trypanosoma brucei development in the tsetse fly vector culminates when parasites gain mammalian infectivity in the salivary glands. A key step in this process is the establishment of monoallelic variant surface glycoprotein (VSG) expression and the formation of the VSG coat. The establishment of VSG monoallelic expression is complex and poorly understood, due to the multiple parasite stages present in the salivary glands. Therefore, we sought to further our understanding of this phenomenon by performing single-cell RNA-sequencing (scRNA-seq) on these trypanosome populations. We were able to capture the developmental program of trypanosomes in the salivary glands, identifying populations of epimastigote, gamete, pre-metacyclic and metacyclic cells. Our results show that parasite metabolism is dramatically remodeled during development in the salivary glands, with a shift in transcript abundance from tricarboxylic acid metabolism to glycolytic metabolism. Analysis of VSG gene expression in pre-metacyclic and metacyclic cells revealed a dynamic VSG gene activation program. Strikingly, we found that pre-metacyclic cells contain transcripts from multiple VSG genes, which resolves to singular VSG gene expression in mature metacyclic cells. Single molecule RNA fluorescence in situ hybridisation (smRNA-FISH) of VSG gene expression following in vitro metacyclogenesis confirmed this finding. Our data demonstrate that multiple VSG genes are transcribed before a single gene is chosen. We propose a transcriptional race model governs the initiation of monoallelic expression.


Subject(s)
Gene Expression Regulation/genetics , Trypanosoma brucei brucei/genetics , Tsetse Flies/parasitology , Variant Surface Glycoproteins, Trypanosoma/genetics , Animals , Insect Vectors/parasitology , RNA-Seq , Salivary Glands/parasitology
11.
Biomolecules ; 11(5)2021 05 11.
Article in English | MEDLINE | ID: mdl-34064869

ABSTRACT

An estimated 229 million people worldwide were impacted by malaria in 2019. The vectors of malaria parasites (Plasmodium spp.) are Anopheles mosquitoes, making their behavior, infection success, and ultimately transmission of great importance. Individuals with severe malaria can exhibit significantly increased blood concentrations of histamine, an allergic mediator in humans and an important insect neuromodulator, potentially delivered to mosquitoes during blood-feeding. To determine whether ingested histamine could alter Anopheles stephensi biology, we provisioned histamine at normal blood levels and at levels consistent with severe malaria and monitored blood-feeding behavior, flight activity, antennal and retinal responses to host stimuli and lifespan of adult female Anopheles stephensi. To determine the effects of ingested histamine on parasite infection success, we quantified midgut oocysts and salivary gland sporozoites in mosquitoes infected with Plasmodium yoelii and Plasmodium falciparum. Our data show that provisioning An. stephensi with histamine at levels consistent with severe malaria can enhance mosquito behaviors and parasite infection success in a manner that would be expected to amplify parasite transmission to and from human hosts. Such knowledge could be used to connect clinical interventions by reducing elevated histamine to mitigate human disease pathology with the delivery of novel lures for improved malaria control.


Subject(s)
Anopheles/drug effects , Histamine/administration & dosage , Malaria/transmission , Mosquito Vectors/drug effects , Plasmodium falciparum/physiology , Salivary Glands/parasitology , Animals , Anopheles/parasitology , Disease Models, Animal , Female , Host-Parasite Interactions , Malaria/parasitology , Malaria/pathology , Mice , Mosquito Vectors/parasitology
12.
Malar J ; 20(1): 284, 2021 Jun 26.
Article in English | MEDLINE | ID: mdl-34174879

ABSTRACT

BACKGROUND: Plasmodium falciparum (Pf) sporozoites (PfSPZ) can be administered as a highly protective vaccine conferring the highest protection seen to date. Sanaria® PfSPZ vaccines are produced using aseptically reared Anopheles stephensi mosquitoes. The bionomics of sporogonic development of P. falciparum in A. stephensi to fully mature salivary gland PfSPZ is thought to be modulated by several components of the mosquito innate immune system. In order to increase salivary gland PfSPZ infections in A. stephensi and thereby increase vaccine production efficiency, a gene knock down approach was used to investigate the activity of the immune deficiency (IMD) signaling pathway downstream effector leucine-rich repeat immune molecule 1 (LRIM1), an antagonist to Plasmodium development. METHODS: Expression of LRIM1 in A. stephensi was reduced following injection of double stranded (ds) RNA into mosquitoes. By combining the Gal4/UAS bipartite system with in vivo expression of short hairpin (sh) RNA coding for LRIM1 reduced expression of LRIM1 was targeted in the midgut, fat body, and salivary glands. RT-qPCR was used to demonstrate fold-changes in gene expression in three transgenic crosses and the effects on P. falciparum infections determined in mosquitoes showing the greatest reduction in LRIM1 expression. RESULTS: LRIM1 expression could be reduced, but not completely silenced, by expression of LRIM1 dsRNA. Infections of P. falciparum oocysts and PfSPZ were consistently and significantly higher in transgenic mosquitoes than wild type controls, with increases in PfSPZ ranging from 2.5- to tenfold. CONCLUSIONS: Plasmodium falciparum infections in A. stephensi can be increased following reduced expression of LRIM1. These data provide the springboard for more precise knockout of LRIM1 for the eventual incorporation of immune-compromised A. stephensi into manufacturing of Sanaria's PfSPZ products.


Subject(s)
Anopheles/parasitology , Insect Proteins/genetics , Plasmodium falciparum/physiology , RNA Interference , Animals , Anopheles/genetics , Female , Gene Knockdown Techniques , Insect Proteins/metabolism , Salivary Glands/parasitology , Sporozoites/physiology
13.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33653959

ABSTRACT

Despite the critical role of Plasmodium sporozoites in malaria transmission, we still know little about the mechanisms underlying their development in mosquitoes. Here, we use single-cell RNA sequencing to characterize the gene expression profiles of 16,038 Plasmodium berghei sporozoites isolated throughout their development from midgut oocysts to salivary glands, and from forced salivation experiments. Our results reveal a succession of tightly regulated changes in gene expression occurring during the maturation of sporozoites and highlight candidate genes that could play important roles in oocyst egress, sporozoite motility, and the mechanisms underlying the invasion of mosquito salivary glands and mammalian hepatocytes. In addition, the single-cell data reveal extensive transcriptional heterogeneity among parasites isolated from the same anatomical site, suggesting that Plasmodium development in mosquitoes is asynchronous and regulated by intrinsic as well as environmental factors. Finally, our analyses show a decrease in transcriptional activity preceding the translational repression observed in mature sporozoites and associated with their quiescent state in salivary glands, followed by a rapid reactivation of the transcriptional machinery immediately upon salivation.


Subject(s)
Anopheles/parasitology , Gene Expression Regulation , Plasmodium berghei/metabolism , Salivary Glands/parasitology , Sporozoites/metabolism , Transcription, Genetic , Animals , Mice
14.
PLoS Negl Trop Dis ; 15(2): e0009071, 2021 02.
Article in English | MEDLINE | ID: mdl-33529215

ABSTRACT

African sleeping sickness is caused by Trypanosoma brucei, a parasite transmitted by the bite of a tsetse fly. Trypanosome infection induces a severe transcriptional downregulation of tsetse genes encoding for salivary proteins, which reduces its anti-hemostatic and anti-clotting properties. To better understand trypanosome transmission and the possible role of glycans in insect bloodfeeding, we characterized the N-glycome of tsetse saliva glycoproteins. Tsetse salivary N-glycans were enzymatically released, tagged with either 2-aminobenzamide (2-AB) or procainamide, and analyzed by HILIC-UHPLC-FLR coupled online with positive-ion ESI-LC-MS/MS. We found that the N-glycan profiles of T. brucei-infected and naïve tsetse salivary glycoproteins are almost identical, consisting mainly (>50%) of highly processed Man3GlcNAc2 in addition to several other paucimannose, high mannose, and few hybrid-type N-glycans. In overlay assays, these sugars were differentially recognized by the mannose receptor and DC-SIGN C-type lectins. We also show that salivary glycoproteins bind strongly to the surface of transmissible metacyclic trypanosomes. We suggest that although the repertoire of tsetse salivary N-glycans does not change during a trypanosome infection, the interactions with mannosylated glycoproteins may influence parasite transmission into the vertebrate host.


Subject(s)
Glycoproteins/metabolism , Lectins, C-Type/metabolism , Polysaccharides/metabolism , Salivary Proteins and Peptides/metabolism , Trypanosoma/metabolism , Tsetse Flies/parasitology , Animals , Chromatography, Liquid , Concanavalin A , Glycoside Hydrolases , Insect Vectors/parasitology , Saliva , Salivary Glands/parasitology , Tandem Mass Spectrometry , Trypanosoma brucei brucei/genetics , Trypanosomiasis, African/parasitology
15.
Malar J ; 20(1): 71, 2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33546698

ABSTRACT

BACKGROUND: Plasmodium falciparum causes the deadliest form of malaria, which remains one of the most prevalent infectious diseases. Unfortunately, the only licensed vaccine showed limited protection and resistance to anti-malarial drug is increasing, which can be largely attributed to the biological complexity of the parasite's life cycle. The progression from one developmental stage to another in P. falciparum involves drastic changes in gene expressions, where its infectivity to human hosts varies greatly depending on the stage. Approaches to identify candidate genes that are responsible for the development of infectivity to human hosts typically involve differential gene expression analysis between stages. However, the detection may be limited to annotated proteins and open reading frames (ORFs) predicted using restrictive criteria. METHODS: The above problem is particularly relevant for P. falciparum; whose genome annotation is relatively incomplete given its clinical significance. In this work, systems proteogenomics approach was used to address this challenge, as it allows computational detection of unannotated, novel Open Reading Frames (nORFs), which are neglected by conventional analyses. Two pairs of transcriptome/proteome were obtained from a previous study where one was collected in the mosquito-infectious oocyst sporozoite stage, and the other in the salivary gland sporozoite stage with human infectivity. They were then re-analysed using the proteogenomics framework to identify nORFs in each stage. RESULTS: Translational products of nORFs that map to antisense, intergenic, intronic, 3' UTR and 5' UTR regions, as well as alternative reading frames of canonical proteins were detected. Some of these nORFs also showed differential expression between the two life cycle stages studied. Their regulatory roles were explored through further bioinformatics analyses including the expression regulation on the parent reference genes, in silico structure prediction, and gene ontology term enrichment analysis. CONCLUSION: The identification of nORFs in P. falciparum sporozoites highlights the biological complexity of the parasite. Although the analyses are solely computational, these results provide a starting point for further experimental validation of the existence and functional roles of these nORFs.


Subject(s)
Anopheles/parasitology , Open Reading Frames , Plasmodium falciparum/genetics , Animals , Computer Simulation , Oocytes/physiology , Proteogenomics , Salivary Glands/parasitology , Sporozoites/physiology
16.
PLoS Comput Biol ; 17(2): e1008658, 2021 02.
Article in English | MEDLINE | ID: mdl-33591963

ABSTRACT

During sporogony, malaria-causing parasites infect a mosquito, reproduce and migrate to the mosquito salivary glands where they can be transmitted the next time blood feeding occurs. The time required for sporogony, known as the extrinsic incubation period (EIP), is an important determinant of malaria transmission intensity. The EIP is typically estimated as the time for a given percentile, x, of infected mosquitoes to develop salivary gland sporozoites (the infectious parasite life stage), which is denoted by EIPx. Many mechanisms, however, affect the observed sporozoite prevalence including the human-to-mosquito transmission probability and possibly differences in mosquito mortality according to infection status. To account for these various mechanisms, we present a mechanistic mathematical model, which explicitly models key processes at the parasite, mosquito and observational scales. Fitting this model to experimental data, we find greater variation in the EIP than previously thought: we estimated the range between EIP10 and EIP90 (at 27°C) as 4.5 days compared to 0.9 days using existing statistical methods. This pattern holds over the range of study temperatures included in the dataset. Increasing temperature from 21°C to 34°C decreased the EIP50 from 16.1 to 8.8 days. Our work highlights the importance of mechanistic modelling of sporogony to (1) improve estimates of malaria transmission under different environmental conditions or disease control programs and (2) evaluate novel interventions that target the mosquito life stages of the parasite.


Subject(s)
Malaria, Falciparum/transmission , Plasmodium falciparum , Salivary Glands/parasitology , Sporozoites/metabolism , Algorithms , Animals , Anopheles , Computer Simulation , Humans , Infectious Disease Incubation Period , Models, Theoretical , Mosquito Vectors/parasitology , Prevalence , Temperature , Time Factors
17.
Malar J ; 20(1): 11, 2021 Jan 06.
Article in English | MEDLINE | ID: mdl-33407511

ABSTRACT

BACKGROUND: The invasion of the mosquito salivary glands by Plasmodium sporozoites is a critical step that defines the success of malaria transmission and a detailed understanding of the molecules responsible for salivary gland invasion could be leveraged towards control of vector-borne pathogens. Antibodies directed against the mosquito salivary gland protein SGS1 have been shown to reduce Plasmodium gallinaceum sporozoite invasion of Aedes aegypti salivary glands, but the specific role of this protein in sporozoite invasion and in other stages of the Plasmodium life cycle remains unknown. METHODS: RNA interference and CRISPR/Cas9 were used to evaluate the role of A. aegypti SGS1 in the P. gallinaceum life cycle. RESULTS: Knockdown and knockout of SGS1 disrupted sporozoite invasion of the salivary gland. Interestingly, mosquitoes lacking SGS1 also displayed fewer oocysts. Proteomic analyses confirmed the abolishment of SGS1 in the salivary gland of SGS1 knockout mosquitoes and revealed that the C-terminus of the protein is absent in the salivary gland of control mosquitoes. In silico analyses indicated that SGS1 contains two potential internal cleavage sites and thus might generate three proteins. CONCLUSION: SGS1 facilitates, but is not essential for, invasion of A. aegypti salivary glands by P. gallinaceum and has a dual role as a facilitator of parasite development in the mosquito midgut. SGS1 could, therefore, be part of a strategy to decrease malaria transmission by the mosquito vector, for example in a transgenic mosquito that blocks its interaction with the parasite.


Subject(s)
Aedes/genetics , Insect Proteins/genetics , Plasmodium gallinaceum/physiology , Salivary Proteins and Peptides/genetics , Aedes/parasitology , Amino Acid Sequence , Animals , Female , Gastrointestinal Tract/parasitology , Insect Proteins/chemistry , Insect Proteins/metabolism , Mosquito Vectors/genetics , Mosquito Vectors/parasitology , Salivary Glands/parasitology , Salivary Proteins and Peptides/chemistry , Salivary Proteins and Peptides/metabolism , Sequence Alignment , Sporozoites/physiology
18.
Parasit Vectors ; 13(1): 441, 2020 Sep 03.
Article in English | MEDLINE | ID: mdl-32883363

ABSTRACT

BACKGROUND: Sand flies are vectors of Leishmania spp., the causative agents of leishmaniasis in vertebrates, including man. The sand fly saliva contains powerful pharmacologically active substances that prevent hemostasis and enhance Leishmania spp. infections. On the other hand, salivary proteins can protect vaccinated mice challenged with parasites. Therefore, sand fly salivary proteins are relevant for the epidemiology of leishmaniasis and can be a potential target for a vaccine against leishmaniasis. Despite this, studies on sand fly salivary glands (SGs) are limited. METHODS: The present study analyzes, in detail, the morphology, anatomy and ultrastructure of the SGs of sand fly vectors of the genera Lutzomyia and Phlebotomus. We used histology, transmission and scanning electron microscopy and lectin labeling associated with confocal laser microscopy. RESULTS: The SGs have conserved and distinct morphological aspects according to the distinct sand fly species. Each SG has a single rounded lobe constituting of c.100-120 secretory cells. The SG secretory cells, according to their ultrastructure and lectin binding, were classified into five different subpopulations, which may differ in secretory pathways. CONCLUSIONS: To the best of our knowledge, these morphological details of sand fly salivary glands are described for the first time. Further studies are necessary to better understand the role of these different cell types and better relate them with the production and secretion of the saliva substances, which has a fundamental role in the interaction of the sand fly vectors with Leishmania.


Subject(s)
Psychodidae/ultrastructure , Salivary Glands/ultrastructure , Animals , Disease Vectors , Leishmaniasis/transmission , Microscopy, Electron , Mosquito Vectors/anatomy & histology , Mosquito Vectors/parasitology , Mosquito Vectors/ultrastructure , Phlebotomus/anatomy & histology , Phlebotomus/parasitology , Phlebotomus/ultrastructure , Psychodidae/anatomy & histology , Psychodidae/parasitology , Salivary Glands/parasitology
19.
mSphere ; 5(4)2020 08 19.
Article in English | MEDLINE | ID: mdl-32817376

ABSTRACT

In the Plasmodium life cycle, two infectious stages of parasites, merozoites and sporozoites, share rhoptry and microneme apical structures. A crucial step during merozoite invasion of erythrocytes is the discharge to the host cell membrane of some rhoptry neck proteins as a complex, followed by the formation of a moving junction involving the parasite-secreted protein AMA1 on the parasite membrane. Components of the merozoite rhoptry neck protein complex are also expressed in sporozoites, namely, RON2, RON4, and RON5, suggesting that invasion mechanism elements might be conserved between these infective stages. Recently, we demonstrated that RON2 is required for sporozoite invasion of mosquito salivary gland cells and mammalian hepatocytes, using a sporozoite stage-specific gene knockdown strategy in the rodent malaria parasite model, Plasmodium berghei Here, we use a coimmunoprecipitation assay and oocyst-derived sporozoite extracts to demonstrate that RON2, RON4, and RON5 also form a complex in sporozoites. The sporozoite stage-specific gene knockdown strategy revealed that both RON4 and RON5 have crucial roles during sporozoite invasion of salivary glands, including a significantly reduced attachment ability required for the onset of gliding. Further analyses indicated that RON2 and RON4 reciprocally affect trafficking to rhoptries in developing sporozoites, while RON5 is independently transported. These findings indicate that the interaction between RON2 and RON4 contributes to their stability and trafficking to rhoptries, in addition to involvement in sporozoite attachment.IMPORTANCE Sporozoites are the motile infectious stage that mediates malaria parasite transmission from mosquitoes to the mammalian host. This study addresses the question whether the rhoptry neck protein complex forms and functions in sporozoites, in addition to its role in merozoites. By applying coimmunoprecipitation and sporozoite stage-specific gene knockdown assays, it was demonstrated that RON2, RON4, and RON5 form a complex and are involved in sporozoite invasion of salivary glands via their attachment ability. These findings shed light on the conserved invasion mechanisms among apicomplexan infective stages. In addition, the sporozoite stage-specific gene knockdown system has revealed for the first time in Plasmodium that the RON2 and RON4 interaction reciprocally affects their stability and trafficking to rhoptries. Our study raises the possibility that the RON complex functions during sporozoite maturation as well as migration toward and invasion of target cells.


Subject(s)
Host-Parasite Interactions , Plasmodium berghei/chemistry , Protozoan Proteins/chemistry , Salivary Glands/parasitology , Sporozoites/physiology , Animals , Culicidae/parasitology , Female , Mice , Mice, Inbred ICR , Plasmodium berghei/genetics , Plasmodium berghei/physiology , Protein Transport , Protozoan Proteins/genetics , Rats , Sporozoites/chemistry
20.
Elife ; 92020 07 10.
Article in English | MEDLINE | ID: mdl-32648541

ABSTRACT

Inserted (I) domains function as ligand-binding domains in adhesins that support cell adhesion and migration in many eukaryotic phyla. These adhesins include integrin αß heterodimers in metazoans and single subunit transmembrane proteins in apicomplexans such as TRAP in Plasmodium and MIC2 in Toxoplasma. Here we show that the I domain of TRAP is essential for sporozoite gliding motility, mosquito salivary gland invasion and mouse infection. Its replacement with the I domain from Toxoplasma MIC2 fully restores tissue invasion and parasite transmission, while replacement with the aX I domain from human integrins still partially restores liver infection. Mutations around the ligand binding site allowed salivary gland invasion but led to inefficient transmission to the rodent host. These results suggest that apicomplexan parasites appropriated polyspecific I domains in part for their ability to engage with multiple ligands and to provide traction for emigration into diverse organs in distant phyla.


Subject(s)
Anopheles/parasitology , Malaria/parasitology , Plasmodium berghei/genetics , Protozoan Proteins/genetics , Amino Acid Sequence , Animals , Evolution, Molecular , Female , Ligands , Mice , Mice, Inbred C57BL , Plasmodium berghei/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Salivary Glands/parasitology , Sequence Alignment , Sporozoites/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...