Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
PLoS Biol ; 20(1): e3001515, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35025886

RESUMO

Anopheles gambiae melanization-based refractoriness to the human malaria parasite Plasmodium falciparum has rarely been observed in either laboratory or natural conditions, in contrast to the rodent model malaria parasite Plasmodium berghei that can become completely melanized by a TEP1 complement-like system-dependent mechanism. Multiple studies have shown that the rodent parasite evades this defense by recruiting the C-type lectins CTL4 and CTLMA2, while permissiveness to the human malaria parasite was not affected by partial depletion of these factors by RNAi silencing. Using CRISPR/Cas9-based CTL4 knockout, we show that A. gambiae can mount melanization-based refractoriness to the human malaria parasite, which is independent of the TEP1 complement-like system and the major anti-Plasmodium immune pathway Imd. Our study indicates a hierarchical specificity in the control of Plasmodium melanization and proves CTL4 as an essential host factor for P. falciparum transmission and one of the most potent mosquito-encoded malaria transmission-blocking targets.


Assuntos
Anopheles/imunologia , Lectinas Tipo C/genética , Plasmodium berghei/fisiologia , Plasmodium falciparum/fisiologia , Animais , Anopheles/genética , Anopheles/parasitologia , Sistemas CRISPR-Cas , Técnicas de Inativação de Genes , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Lectinas Tipo C/metabolismo , Melaninas/genética , Melaninas/imunologia
2.
J Infect Dis ; 216(10): 1264-1272, 2017 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-28968664

RESUMO

Background: Complete malaria eradication and optimal use of transmission-reducing interventions require knowledge of submicroscopic infectious reservoirs among asymptomatic individuals. Even submicroscopic levels of Plasmodium falciparum gametocytes can infect mosquitoes and promote onward transmission. Most efforts to identify gametocyte carriers use polymerase chain reaction amplification of the gametocyte-specific transcript Pfs25. Methods: To expand the repertoire of biomarkers available for superior gametocyte detection, we compared the gene expression profiles of gametocytes and asynchronous blood-stage P. falciparum parasites by microarray technology. This allowed the identification of 56 molecules abundantly expressed in the gametocyte stage of the parasite. The analytical sensitivity for gametocyte detection was evaluated for 25 genes with the highest expression levels. Results: One candidate, Pfg17, exhibited superior analytical sensitivity against a panel of gametocyte-spiked whole blood, detecting 10 gametocytes/mL; in comparison, Pfs25 detected only 25.3 gametocytes/mL. Pfg17 also exhibited superior clinical sensitivity, identifying 19.1% more samples from blood-film microscopy-negative Ghanaian children and 40% more samples from asymptomatic adults as gametocyte positive. Conclusions: Cumulatively, our results suggest Pfg17 is an excellent biomarker for detecting asymptomatic infectious reservoirs otherwise missed by the most sensitive molecular method available. Our study has also improved the repertoire of transmission-stage antigens available for evaluation as candidate vaccines.


Assuntos
Reservatórios de Doenças/parasitologia , Malária Falciparum/parasitologia , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Adolescente , Biomarcadores , Criança , Pré-Escolar , Feminino , Perfilação da Expressão Gênica , Genes de Protozoários , Humanos , Lactente , Recém-Nascido , Malária Falciparum/epidemiologia , Malária Falciparum/transmissão , Masculino , Parasitemia/parasitologia , Reação em Cadeia da Polimerase/métodos , Sensibilidade e Especificidade
3.
Biochem Biophys Res Commun ; 493(1): 690-696, 2017 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-28864420

RESUMO

K+ channels are integral membrane proteins, which contribute to maintain vital parameters such as the cellular membrane potential and cell volume. Malaria parasites encode two K+ channel homologues, Kch1 and Kch2, which are well-conserved among members of the Plasmodium genus. In the rodent malaria parasite P. berghei, the functional significance of K+ channel homologue PbKch2 was studied using targeted gene knock-out. The knockout parasites were characterized in a mouse model in terms of growth-kinetics and infectivity in the mosquito vector. Furthermore, using a tracer-uptake technique with 86Rb+ as a K+ congener, the K+ transporting properties of the knockout parasites were assessed. RESULTS: Genetic disruption of Kch2 did not grossly affect the phenotype in terms of asexual replication and pathogenicity in a mouse model. In contrast to Kch1-null parasites, Kch2-null parasites were fully capable of forming oocysts in female Anopheles stephensi mosquitoes. 86Rb+ uptake in Kch2-deficient blood-stage P. berghei parasites (Kch2-null) did not differ from that of wild-type (WT) parasites. About two-thirds of the 86Rb+ uptake in WT and in Kch2-null parasites could be inhibited by K+ channel blockers and could be inferred to the presence of functional Kch1 in Kch2 knockout parasites. Kch2 is therefore not required for transport of K+ in P. berghei and is not essential to mosquito-stage sporogonic development of the parasite.


Assuntos
Anopheles/parasitologia , Malária/parasitologia , Plasmodium berghei/metabolismo , Plasmodium berghei/patogenicidade , Canais de Potássio/metabolismo , Proteínas de Protozoários/metabolismo , Animais , Feminino , Masculino , Camundongos , Plasmodium berghei/genética , Canais de Potássio/genética , Proteínas de Protozoários/genética
4.
PLoS Pathog ; 10(10): e1004398, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25340821

RESUMO

Plasmodium and dengue virus, the causative agents of the two most devastating vector-borne diseases, malaria and dengue, are transmitted by the two most important mosquito vectors, Anopheles gambiae and Aedes aegypti, respectively. Insect-bacteria associations have been shown to influence vector competence for human pathogens through multi-faceted actions that include the elicitation of the insect immune system, pathogen sequestration by microbes, and bacteria-produced anti-pathogenic factors. These influences make the mosquito microbiota highly interesting from a disease control perspective. Here we present a bacterium of the genus Chromobacterium (Csp_P), which was isolated from the midgut of field-caught Aedes aegypti. Csp_P can effectively colonize the mosquito midgut when introduced through an artificial nectar meal, and it also inhibits the growth of other members of the midgut microbiota. Csp_P colonization of the midgut tissue activates mosquito immune responses, and Csp_P exposure dramatically reduces the survival of both the larval and adult stages. Ingestion of Csp_P by the mosquito significantly reduces its susceptibility to Plasmodium falciparum and dengue virus infection, thereby compromising the mosquito's vector competence. This bacterium also exerts in vitro anti-Plasmodium and anti-dengue activities, which appear to be mediated through Csp_P -produced stable bioactive factors with transmission-blocking and therapeutic potential. The anti-pathogen and entomopathogenic properties of Csp_P render it a potential candidate for the development of malaria and dengue control strategies.


Assuntos
Anopheles/microbiologia , Chromobacterium/metabolismo , Vírus da Dengue , Infecções por Bactérias Gram-Negativas/metabolismo , Malária/microbiologia , Animais , Culicidae , Vetores Genéticos/genética , Humanos , Técnicas In Vitro , Plasmodium falciparum/microbiologia , Fatores de Virulência
5.
Malar J ; 15(1): 425, 2016 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-27549662

RESUMO

BACKGROUND: Malaria exerts a tremendous socioeconomic impact worldwide despite current control efforts, and novel disease transmission-blocking strategies are urgently needed. The Enterobacter bacterium Esp_Z, which is naturally harboured in the mosquito midgut, can inhibit the development of Plasmodium parasites prior to their invasion of the midgut epithelium through a mechanism that involves oxidative stress. Here, a multifaceted approach is used to study the tripartite interactions between the mosquito, Esp_Z and Plasmodium, towards addressing the feasibility of using sugar-baited exposure of mosquitoes to the Esp_Z bacterium for interruption of malaria transmission. METHODS: The ability of Esp_Z to colonize Anopheles gambiae midguts harbouring microbiota derived from wild mosquitoes was determined by qPCR. Upon introduction of Esp_Z via nectar feeding, the permissiveness of colonized mosquitoes to Plasmodium falciparum infection was determined, as well as the impact of Esp_Z on mosquito fitness parameters, such as longevity, number of eggs laid and number of larvae hatched. The genome of Esp_Z was sequenced, and transcriptome analyses were performed to identify bacterial genes that are important for colonization of the mosquito midgut, as well as for ROS-production. A gene expression analysis of members of the oxidative defence pathway of Plasmodium berghei was also conducted to assess the parasite's oxidative defence response to Esp_Z exposure. RESULTS: Esp_Z persisted for up to 4 days in the An. gambiae midgut after introduction via nectar feeding, and was able to significantly inhibit Plasmodium sporogonic development. Introduction of this bacterium did not adversely affect mosquito fitness. Candidate genes involved in the selection of a better fit Esp_Z to the mosquito midgut environment and in its ability to condition oxidative status of its surroundings were identified, and parasite expression data indicated that Esp_Z is able to induce a partial and temporary shutdown of the ookinetes antioxidant response. CONCLUSIONS: Esp_Z is capable of inhibiting sporogonic development of Plasmodium in the presence of the mosquito's native microbiota without affecting mosquito fitness. Several candidate bacterial genes are likely mediating midgut colonization and ROS production, and inhibition of Plasmodium development appears to involve a shutdown of the parasite's oxidative defence system. A better understanding of the complex reciprocal tripartite interactions can facilitate the development and optimization of an Esp_Z-based malaria control strategy.


Assuntos
Anopheles/microbiologia , Anopheles/parasitologia , Enterobacter/crescimento & desenvolvimento , Interações Microbianas , Mosquitos Vetores/microbiologia , Mosquitos Vetores/parasitologia , Plasmodium/crescimento & desenvolvimento , Animais , Enterobacter/genética , Feminino , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/parasitologia , Longevidade , Oviposição , Plasmodium/genética , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sobrevida
6.
Environ Microbiol ; 16(9): 2980-94, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24428613

RESUMO

Malaria parasite transmission requires the successful development of Plasmodium gametocytes into flagellated microgametes upon mosquito blood ingestion, and the subsequent fertilization of microgametes and macrogametes for the development of motile zygotes, called ookinetes, which invade and transverse the Anopheles vector mosquito midgut at around 18-36 h after blood ingestion. Within the mosquito midgut, the malaria parasite has to withstand the mosquito's innate immune response and the detrimental effect of its commensal bacterial flora. We have assessed the midgut colonization capacity of five gut bacterial isolates from field-derived, and two from laboratory colony, mosquitoes and their effect on Plasmodium development in vivo and in vitro, along with their impact on mosquito survival. Some bacterial isolates activated the mosquito's immune system, affected the mosquito's lifespan, and were capable of blocking Plasmodium development. We have also shown that the ability of these bacteria to inhibit the parasites is likely to involve different mechanisms and factors. A Serratia marcescens isolate was particularly efficient in colonizing the mosquitoes' gut, compromising mosquito survival and inhibiting both Plasmodium sexual- and asexual-stage through secreted factors, thereby rendering it a potential candidate for the development of a malaria transmission intervention strategy.


Assuntos
Anopheles/microbiologia , Sistema Digestório/microbiologia , Plasmodium/microbiologia , Serratia marcescens/fisiologia , Animais , Anopheles/imunologia , Anopheles/parasitologia , Bactérias/isolamento & purificação , Feminino , Imunidade Inata , Camundongos , Serratia marcescens/isolamento & purificação
7.
Genome Res ; 21(11): 1872-81, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21795387

RESUMO

Anopheles gambiae is a major mosquito vector responsible for malaria transmission, whose genome sequence was reported in 2002. Genome annotation is a continuing effort, and many of the approximately 13,000 genes listed in VectorBase for Anopheles gambiae are predictions that have still not been validated by any other method. To identify protein-coding genes of An. gambiae based on its genomic sequence, we carried out a deep proteomic analysis using high-resolution Fourier transform mass spectrometry for both precursor and fragment ions. Based on peptide evidence, we were able to support or correct more than 6000 gene annotations including 80 novel gene structures and about 500 translational start sites. An additional validation by RT-PCR and cDNA sequencing was successfully performed for 105 selected genes. Our proteogenomic analysis led to the identification of 2682 genome search-specific peptides. Numerous cases of encoded proteins were documented in regions annotated as intergenic, introns, or untranslated regions. Using a database created to contain potential splice sites, we also identified 35 novel splice junctions. This is a first report to annotate the An. gambiae genome using high-accuracy mass spectrometry data as a complementary technology for genome annotation.


Assuntos
Anopheles/genética , Anopheles/metabolismo , Processamento Alternativo , Animais , Mapeamento Cromossômico , Códon de Iniciação , Éxons , Genes de Insetos , Genômica , Íntrons , Espectrometria de Massas , Anotação de Sequência Molecular , Dados de Sequência Molecular , Fases de Leitura Aberta , Peptídeos/genética , Proteômica , Sítios de Splice de RNA , Reprodutibilidade dos Testes , Regiões não Traduzidas/genética
8.
bioRxiv ; 2023 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-36798298

RESUMO

Rising numbers of malaria cases and deaths underscore the need for new interventions. Long-acting injectable medications, such as those now in use for HIV prophylaxis, offer the prospect of a malaria "chemical vaccine", combining the efficacy of a drug (like atovaquone) with the durability of a biological vaccine. Of concern, however, is the possible selection and transmission of drug-resistant parasites. We addressed this question by generating clinically relevant, highly atovaquone-resistant, Plasmodium falciparum mutants competent to infect mosquitoes. Isogenic paired strains, that differ only by a single Y268S mutation in cytochrome b, were evaluated in parallel in southeast Asian (Anopheles stephensi) or African (Anopheles gambiae) mosquitoes, and thence in humanized mice. Fitness costs of the mutation were evident along the lifecycle, in asexual parasite growth in vitro and in a progressive loss of parasites in the mosquito. In numerous independent experiments, microscopic exam of salivary glands from hundreds of mosquitoes failed to detect even one Y268S sporozoite, a defect not rescued by coinfection with wild type parasites. Furthermore, despite uniformly successful transmission of wild type parasites from An. stephensi to FRG NOD huHep mice bearing human hepatocytes and erythrocytes, multiple attempts with Y268S-fed mosquitoes failed: there was no evidence of parasites in mouse tissues by microscopy, in vitro culture, or PCR. These studies confirm a severe-to-lethal fitness cost of clinically relevant atovaquone-resistant P. falciparum in the mosquito, and they significantly lessen the likelihood of their transmission in the field.

9.
Nat Commun ; 14(1): 6415, 2023 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-37828012

RESUMO

Long-acting injectable medications, such as atovaquone, offer the prospect of a "chemical vaccine" for malaria, combining drug efficacy with vaccine durability. However, selection and transmission of drug-resistant parasites is of concern. Laboratory studies have indicated that atovaquone resistance disadvantages parasites in mosquitoes, but lack of data on clinically relevant Plasmodium falciparum has hampered integration of these variable findings into drug development decisions. Here we generate atovaquone-resistant parasites that differ from wild type parent by only a Y268S mutation in cytochrome b, a modification associated with atovaquone treatment failure in humans. Relative to wild type, Y268S parasites evidence multiple defects, most marked in their development in mosquitoes, whether from Southeast Asia (Anopheles stephensi) or Africa (An. gambiae). Growth of asexual Y268S P. falciparum in human red cells is impaired, but parasite loss in the mosquito is progressive, from reduced gametocyte exflagellation, to smaller number and size of oocysts, and finally to absence of sporozoites. The Y268S mutant fails to transmit from mosquitoes to mice engrafted with human liver cells and erythrocytes. The severe-to-lethal fitness cost of clinically relevant atovaquone resistance to P. falciparum in the mosquito substantially lessens the likelihood of its transmission in the field.


Assuntos
Anopheles , Antimaláricos , Malária Falciparum , Malária , Parasitos , Vacinas , Humanos , Animais , Camundongos , Atovaquona/farmacologia , Atovaquona/uso terapêutico , Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , Malária/parasitologia , Malária Falciparum/tratamento farmacológico , Plasmodium falciparum/genética , Anopheles/parasitologia , Antiparasitários/uso terapêutico
10.
bioRxiv ; 2023 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-37398288

RESUMO

Drug-resistant Plasmodium falciparum parasites have swept across Southeast Asia and now threaten Africa. By implementing a P. falciparum genetic cross using humanized mice, we report the identification of key determinants of resistance to artemisinin (ART) and piperaquine (PPQ) in the dominant Asian KEL1/PLA1 lineage. We mapped k13 as the central mediator of ART resistance and identified secondary markers. Applying bulk segregant analysis, quantitative trait loci mapping and gene editing, our data reveal an epistatic interaction between mutant PfCRT and multicopy plasmepsins 2/3 in mediating high-grade PPQ resistance. Susceptibility and parasite fitness assays implicate PPQ as a driver of selection for KEL1/PLA1 parasites. Mutant PfCRT enhanced susceptibility to lumefantrine, the first-line partner drug in Africa, highlighting a potential benefit of opposing selective pressures with this drug and PPQ. We also identified that the ABCI3 transporter can operate in concert with PfCRT and plasmepsins 2/3 in mediating multigenic resistance to antimalarial agents.

11.
Sci Adv ; 9(45): eadi2364, 2023 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-37939186

RESUMO

Drug-resistant Plasmodium falciparum parasites have swept across Southeast Asia and now threaten Africa. By implementing a P. falciparum genetic cross using humanized mice, we report the identification of key determinants of resistance to artemisinin (ART) and piperaquine (PPQ) in the dominant Asian KEL1/PLA1 lineage. We mapped k13 as the central mediator of ART resistance in vitro and identified secondary markers. Applying bulk segregant analysis, quantitative trait loci mapping using 34 recombinant haplotypes, and gene editing, our data reveal an epistatic interaction between mutant PfCRT and multicopy plasmepsins 2/3 in mediating high-grade PPQ resistance. Susceptibility and parasite fitness assays implicate PPQ as a driver of selection for KEL1/PLA1 parasites. Mutant PfCRT enhanced susceptibility to lumefantrine, the first-line partner drug in Africa, highlighting a potential benefit of opposing selective pressures with this drug and PPQ. We also identified that the ABCI3 transporter can operate in concert with PfCRT and plasmepsins 2/3 in mediating multigenic resistance to antimalarial agents.


Assuntos
Malária Falciparum , Parasitos , Animais , Camundongos , Plasmodium falciparum/genética , Malária Falciparum/tratamento farmacológico , Malária Falciparum/genética , Malária Falciparum/parasitologia , Resistência a Medicamentos/genética , Resistência a Múltiplos Medicamentos , Genômica
12.
Proc Natl Acad Sci U S A ; 105(17): 6398-402, 2008 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-18434537

RESUMO

Regulated K(+) transport across the plasma membrane is of vital importance for the survival of most cells. Two K(+) channels have been identified in the Plasmodium falciparum genome; however, their functional significance during parasite life cycle in the vertebrate host and during transmission through the mosquito vector remains unknown. We hypothesize that these two K(+) channels mediate the transport of K(+) in the parasites, and thus are important for parasite survival. To test this hypothesis, we identified the orthologue of one of the P. falciparum K(+) channels, PfKch1, in the rodent malaria parasite P. berghei (PbKch1) and examined the biological role by performing a targeted disruption of the gene encoding PbKch1. The deduced amino acid sequence of the six transmembrane domains of PfKch1 and PbKch1 share 82% identity, and in particular the pore regions are completely identical. The PbKch1-null parasites were viable despite a marked reduction in the uptake of the K(+) congener (86)Rb(+), and mice infected with PbKch1-null parasites survived slightly longer than mice infected with WT parasites. However, the most striking feature of the phenotype was the virtually complete inhibition of the development of PbKch1-null parasites in Anopheles stephensi mosquitoes. In conclusion, these studies demonstrate that PbKch1 contributes to the transport of K(+) in P. berghei parasites and supports the growth of the parasites, in particular the development of oocysts in the mosquito midgut. K(+) channels therefore may constitute a potential antimalarial drug target.


Assuntos
Marcação de Genes , Malária/transmissão , Plasmodium berghei/genética , Canais de Potássio/genética , Sequência de Aminoácidos , Animais , Anopheles/parasitologia , Cinética , Estágios do Ciclo de Vida , Malária/parasitologia , Camundongos , Dados de Sequência Molecular , Parasitos/genética , Parasitos/crescimento & desenvolvimento , Parasitos/patogenicidade , Fenótipo , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/patogenicidade , Plasmodium falciparum , Potássio/metabolismo , Canais de Potássio/química , Rubídio/metabolismo , Análise de Sequência de Proteína , Homologia de Sequência de Aminoácidos , Virulência
13.
NPJ Vaccines ; 6(1): 98, 2021 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-34376691

RESUMO

Genetically attenuated sporozoite vaccines can elicit long-lasting protection against malaria but pose risks of breakthrough infection. Chemoprophylaxis vaccination (CVac) has proven to be the most effective vaccine strategy against malaria. Here, we demonstrate that a liver stage-specific autophagy mutant of Plasmodium berghei (ATG8 overexpressor), when used as a live vaccine under a CVac regimen, provides superior long-lasting protection, in both inbred and outbred mice, as compared to WT-CVac. Uniquely, the protection elicited by this mutant is predominantly dependent on a CD8+ T-cell response through an IFN-γ-independent mechanism and is associated with a stable population of antigen-experienced CD8+ T cells. Jointly, our findings support the exploitation of liver-stage mutants as vaccines under a CVac protocol. This vaccination strategy is also a powerful model to study the mechanisms of protective immunity and discover new protective antigens.

14.
Sci Transl Med ; 13(597)2021 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-34108248

RESUMO

Malaria vaccines that disrupt the Plasmodium life cycle in mosquitoes and reduce parasite transmission in endemic areas are termed transmission-blocking vaccines (TBVs). Despite decades of research, there are only a few Plasmodium falciparum antigens that indisputably and reproducibly demonstrate transmission-blocking immunity. So far, only two TBV candidates have advanced to phase 1/2 clinical testing with limited success. By applying an unbiased transcriptomics-based approach, we have identified Pf77 and male development gene 1 (PfMDV-1) as two P. falciparum TBV antigens that, upon immunization, induced antibodies that caused reductions in oocyst counts in Anopheles mosquito midguts in a standard membrane feeding assay. In-depth studies were performed to characterize the genetic diversity of, stage-specific expression by, and natural immunity to these two molecules to evaluate their suitability as TBV candidates. Pf77 and PfMDV-1 display limited antigenic polymorphism, are pan-developmentally expressed within the parasite, and induce naturally occurring antibodies in Ghanaian adults, which raises the prospect of natural boosting of vaccine-induced immune response in endemic regions. Together, these biological properties suggest that Pf77 and PfMDV-1 may warrant further investigation as TBV candidates.


Assuntos
Vacinas Antimaláricas , Malária Falciparum , Animais , Anticorpos Antiprotozoários , Antígenos de Protozoários/genética , Gana , Malária Falciparum/prevenção & controle , Masculino , Plasmodium falciparum
15.
J Vis Exp ; (161)2020 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-32716382

RESUMO

Malaria remains one of the most important public health problems, causing significant morbidity and mortality. Malaria is a mosquito borne disease transmitted through an infectious bite from the female Anopheles mosquito. Malaria control will eventually rely on a multitude of approaches, which includes ways to block transmission to, through and from mosquitoes. To study mosquito stages of malaria parasites in the laboratory, we have optimized a protocol to culture highly infectious Plasmodium falciparum gametocytes, a parasite stage required for transmission from the human host to the mosquito vector. P. falciparum gametocytes mature through five morphologically distinct steps, which takes approximately 1-2 weeks. Gametocyte culture described in this protocol is completed in 15 days and are infectious to mosquitoes from days 15-18. These protocols were developed to maintain a continuous cycle of infection competent gametocytes and to maintain uninterrupted supply of mosquito stages of the parasite. Here, we describe the methodology of gametocyte culture and how to infect mosquitoes with these parasites using glass membrane feeders.


Assuntos
Anopheles/parasitologia , Plasmodium falciparum , Animais , Feminino , Humanos , Malária Falciparum , Membranas Artificiais , Mosquitos Vetores
16.
Sci Rep ; 9(1): 13131, 2019 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-31511546

RESUMO

Malaria parasites have a complex life cycle that includes specialized stages for transmission between their mosquito and human hosts. These stages are an understudied part of the lifecycle yet targeting them is an essential component of the effort to shrink the malaria map. The human parasite Plasmodium falciparum is responsible for the majority of deaths due to malaria. Our goal was to generate transgenic P. falciparum lines that could complete the lifecycle and produce fluorescent transmission stages for more in-depth and high-throughput studies. Using zinc-finger nuclease technology to engineer an integration site, we generated three transgenic P. falciparum lines in which tdtomato or gfp were stably integrated into the genome. Expression was driven by either stage-specific peg4 and csp promoters or the constitutive ef1a promoter. Phenotypic characterization of these lines demonstrates that they complete the life cycle with high infection rates and give rise to fluorescent mosquito stages. The transmission stages are sufficiently bright for intra-vital imaging, flow cytometry and scalable screening of chemical inhibitors and inhibitory antibodies.


Assuntos
Proteínas de Fluorescência Verde/genética , Proteínas Luminescentes/genética , Malária Falciparum/transmissão , Parasitos/genética , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Animais , Culicidae/parasitologia , Citometria de Fluxo/métodos , Engenharia Genética/métodos , Proteínas de Fluorescência Verde/metabolismo , Humanos , Estágios do Ciclo de Vida , Proteínas Luminescentes/metabolismo , Malária Falciparum/parasitologia , Microscopia de Fluorescência/métodos , Parasitos/crescimento & desenvolvimento , Parasitos/fisiologia , Fenótipo , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/metabolismo , Proteína Vermelha Fluorescente
17.
Infect Immun ; 76(5): 2018-24, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18316385

RESUMO

Currently, there is no animal model for Plasmodium falciparum challenge to evaluate malaria transmission-blocking vaccines based on the well-established Pfs25 target antigen. The biological activity of transmission-blocking antibodies is typically assessed using an assay known as the membrane feeding assay (MFA). It is an in vitro method that involves mixing antibodies with cultured P. falciparum gametocytes and feeding them to mosquitoes through an artificial membrane followed by assessment of infection in the mosquitoes. We genetically modified Plasmodium berghei to express Pfs25 and demonstrated that the transgenic parasites (TrPfs25Pb) are susceptible to anti-Pfs25 antibodies during mosquito-stage development. The asexual growth kinetics and mosquito infectivity of TrPfs25Pb were comparable to those of wild-type parasites, and TrPfs25Pb displayed Pfs25 on the surface of ookinetes. Immune sera from nonhuman primates immunized with a Pfs25-based vaccine when passively transferred to mice blocked transmission of TrPfs25Pb to Anopheles stephensi. Furthermore, mice immunized with Pfs25 DNA vaccine and challenged with TrPfs25Pb displayed reduced malaria transmission compared to mice immunized with wild-type plasmid. These studies describe development of an animal malaria model alternative to the in vitro MFA and show that the model can facilitate P. falciparum transmission-blocking vaccine evaluation based on the target antigen Pfs25. We believe that an animal model to test transmission-blocking vaccines would be superior to the MFA, since there may be additional immune factors that synergize the transmission-blocking activity of antibodies in vivo.


Assuntos
Modelos Animais de Doenças , Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Plasmodium berghei/genética , Plasmodium berghei/patogenicidade , Proteínas de Protozoários/imunologia , Animais , Animais Geneticamente Modificados , Anopheles/parasitologia , Anticorpos Antiprotozoários/imunologia , Contagem de Células , Feminino , Malária Falciparum/imunologia , Camundongos , Plasmodium berghei/imunologia , Plasmodium falciparum/imunologia , Esporos de Protozoários
18.
Am J Trop Med Hyg ; 78(1): 114-6, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18187793

RESUMO

Plasmodium falciparum gametocytes are obligate parasite sexual stages required for transmission of malaria from human hosts to the mosquito vector. Assessment of gametocyte carriers in the population is critical in understanding malaria transmission dynamics and in epidemiology studies. We applied a reverse transcription-polymerase chain reaction (RT-PCR)-based approach to detect pfs25 transcripts from blood dried on different filter papers in the laboratory. The detection limit was 1-2 gametocytes/microL. We further validated this assay by analyzing RNA in 10 matched blood samples (liquid blood and blood spotted on filter papers) collected from subjects under field conditions in Zambia. These results thus establish feasibility of detection of Plasmodium falciparum gametocytes by RT-PCR method from dried blood on filter paper. This assay will greatly facilitate bulk analysis of gametocyte RNA transcripts on filter paper, especially in areas where collection and preservation of liquid blood is not feasible.


Assuntos
Filtração , Malária Falciparum/diagnóstico , Plasmodium falciparum/isolamento & purificação , Animais , Primers do DNA , Células Germinativas , Humanos , Malária Falciparum/sangue , Plasmodium falciparum/genética , Valor Preditivo dos Testes , RNA de Protozoário/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Zâmbia
19.
Sci Rep ; 8(1): 655, 2018 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-29330527

RESUMO

The discovery of aquaglyceroporins (AQP) has highlighted a new mechanism of membrane solute transport that may hold therapeutic potential for controlling parasitic infections, including malaria. Plasmodium parasites express a single AQP at the plasma membrane that functions as a channel for water, nutrients and waste into and out cells. We previously demonstrated that Plasmodium berghei targeted for PbAQP deletion are deficient in glycerol import and less virulent than wild-type parasites during the blood developmental stage. Here, we have examined the contribution of PbAQP to the infectivity of P. berghei in the liver. PbAQP is expressed in the sporozoite mosquito stage and is detected at low levels in intrahepatic parasites at the onset of hepatocyte infection. As the parasites progress to late hepatic stages, PbAQP transcription increases and PbAQP localizes to the plasma membrane of hepatic merozoites. Compared to wild-type parasites, PbAQP-null sporozoites exhibit a delay in blood stage infection due to slower replication in hepatocytes, resulting in retardation of merosome production. Furthermore, PbAQP disruption results in a significant reduction in erythrocyte infectivity by hepatocyte-derived merozoites. Hepatic merozoites incorporate exogenous glycerol into glycerophospholipids and PbAQP-null merozoites contain less phosphatidylcholine than wild-type merozoites, underlining the contribution of Plasmodium AQP to phospholipid syntheses.


Assuntos
Aquagliceroporinas/metabolismo , Fígado/parasitologia , Malária/parasitologia , Plasmodium berghei/patogenicidade , Animais , Aquagliceroporinas/genética , Linhagem Celular , Membrana Celular/metabolismo , Eritrócitos/parasitologia , Glicerol/metabolismo , Glicerofosfolipídeos/metabolismo , Camundongos , Plasmodium berghei/metabolismo , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Esporozoítos/metabolismo
20.
Nat Commun ; 9(1): 2908, 2018 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-30046053

RESUMO

The key step during the initiation of malaria is for motile Plasmodium parasites to exit the host dermis and infect the liver. During transmission, the parasites in the form of sporozoites, are injected together with mosquito saliva into the skin. However, the contribution of vector saliva to sporozoite activity during the establishment of the initial infection of the liver is poorly understood. Here we identify a vector protein by mass spectrometry, with similarity to the human gamma interferon inducible thiol reductase (GILT), that is associated with saliva sporozoites of infected Anopheles mosquitoes and has a negative impact on the speed and cell traversal activity of Plasmodium. This protein, referred to as mosquito GILT (mosGILT) represents an example of a protein found in mosquito saliva that may negatively influence sporozoite movement in the host and could lead to new approaches to prevent malaria.


Assuntos
Proteínas de Insetos/metabolismo , Malária/parasitologia , Malária/transmissão , Mosquitos Vetores/parasitologia , Plasmodium/patogenicidade , Glândulas Salivares/parasitologia , Esporozoítos/patogenicidade , Animais , Interações Hospedeiro-Parasita , Proteínas de Insetos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA