Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
Más filtros

Intervalo de año de publicación
1.
PLoS Pathog ; 18(10): e1010887, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36223427

RESUMEN

Plasmodium parasites are reliant on the Apicomplexan AP2 (ApiAP2) transcription factor family to regulate gene expression programs. AP2 DNA binding domains have no homologs in the human or mosquito host genomes, making them potential antimalarial drug targets. Using an in-silico screen to dock thousands of small molecules into the crystal structure of the AP2-EXP (Pf3D7_1466400) AP2 domain (PDB:3IGM), we identified putative AP2-EXP interacting compounds. Four compounds were found to block DNA binding by AP2-EXP and at least one additional ApiAP2 protein. Our top ApiAP2 competitor compound perturbs the transcriptome of P. falciparum trophozoites and results in a decrease in abundance of log2 fold change > 2 for 50% (46/93) of AP2-EXP target genes. Additionally, two ApiAP2 competitor compounds have multi-stage anti-Plasmodium activity against blood and mosquito stage parasites. In summary, we describe a novel set of antimalarial compounds that interact with AP2 DNA binding domains. These compounds may be used for future chemical genetic interrogation of ApiAP2 proteins or serve as starting points for a new class of antimalarial therapeutics.


Asunto(s)
Antimaláricos , Proteínas de Unión al ADN , Plasmodium , Humanos , Antimaláricos/farmacología , Antimaláricos/metabolismo , ADN/metabolismo , Plasmodium/efectos de los fármacos , Plasmodium/genética , Proteínas Protozoarias/metabolismo , Proteínas de Unión al ADN/metabolismo
2.
Cell Tissue Res ; 393(2): 297-320, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37272999

RESUMEN

The mosquito larval midgut is responsible for acquiring and storing most of the nutrients that will sustain the events of metamorphosis and the insect's adult life. Despite its importance, the basic biology of this larval organ is poorly understood. To help fill this gap, we carried out a comparative morphophysiological investigation of three larval midgut regions (gastric caeca, anterior midgut, and posterior midgut) of phylogenetically distant mosquitoes: Anopheles gambiae (Anopheles albimanus was occasionally used as an alternate), Aedes aegypti, and Toxorhynchites theobaldi. Larvae of Toxorhynchites mosquitoes are predacious, in contrast to the other two species, that are detritivorous. In this work, we show that the larval gut of the three species shares basic histological characteristics, but differ in other aspects. The lipid and carbohydrate metabolism of the An. gambiae larval midgut is different compared with that of Ae. aegypti and Tx. theobaldi. The gastric caecum is the most variable region, with differences probably related to the chemical composition of the diet. The peritrophic matrix is morphologically similar in the three species, and processes involved in the post-embryonic development of the organ, such as cell differentiation and proliferation, were also similar. FMRF-positive enteroendocrine cells are grouped in the posterior midgut of Tx. theobaldi, but individualized in An. gambiae and Ae. aegypti. We hypothesize that Tx. theobaldi larval predation is an ancestral condition in mosquito evolution.


Asunto(s)
Aedes , Anopheles , Animales , Anopheles/fisiología , Larva/metabolismo , Sistema Digestivo , Células Enteroendocrinas
3.
Microb Ecol ; 83(3): 766-775, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34231036

RESUMEN

Paratransgenesis consists of genetically engineering an insect symbiont to control vector-borne diseases. Biosafety assessments are a prerequisite for the use of genetically modified organisms (GMOs). Assessments rely on the measurement of the possible impacts of GMOs on different organisms, including beneficial organisms, such as pollinators. The bacterium Serratia AS1 has been genetically modified to express anti-Plasmodium effector proteins and does not impose a fitness cost on mosquitoes that carry it. In the present study, we assessed the impact of this bacterium on the native bee Partamona helleri (Meliponini), an ecologically important species in Brazil. Serratia eGFP AS1 (recombinant strain) or a wild strain of Serratia marcescens were suspended in a sucrose solution and fed to foragers, followed by measurements of survival, feeding rate, and behavior (walking and flying). These bacteria did not change any of the variables measured at 24, 72, and 144 h after the onset of the experiment. Recombinant and wild bacteria were detected in the homogenates of digestive tract during the 144 h period analyzed, but their numbers decreased with time. The recombinant strain was detected in the midgut at 24 h and in the hindgut at 72 h and 144 h after the onset of the experiment under the fluorescent microscope. As reported for mosquitoes, Serratia eGFP AS1 did not compromise the foragers of P. helleri, an ecologically relevant bee.


Asunto(s)
Mosquitos Vectores , Serratia , Animales , Abejas , Brasil , Serratia/genética
4.
Cell Tissue Res ; 385(3): 585-602, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33961128

RESUMEN

FMRFamide-related peptides (FaRPs) are a class of neuropeptides that participate in a variety of physiological processes in invertebrates. They occur in nerves of stomatogastric ganglia and enteroendocrine cells of the insect digestive tract, where they may control muscle functions. However, their direct involvement in muscle function has never been shown in situ. We studied the relationship between FaRPs and midgut muscle during larval-pupal transition of the mosquito Aedes aegypti. In late L4, FaRP-positive neuronal extensions attach to the bundles of the external circular muscle layer, and muscle stem cells start to undergo mitosis in the internal circular layer. Thereafter, the external muscle layer degenerates, disappearing during early pupal development, and is completely absent in the adult mosquito. Our results indicate that FaRP-based neural signals are involved in the reorganization of the muscle fibers of the mosquito midgut during the larval-pupal transition. In addition to confirming FaRP involvement in muscle function, we show that the mosquito midgut muscles are largely innervated, and that circular and longitudinal muscle have specific neuron bodies associated with them.


Asunto(s)
Sistema Nervioso Entérico/fisiología , FMRFamida/metabolismo , Enfermedades Neuromusculares/fisiopatología , Péptidos/metabolismo , Aedes , Animales
5.
BMC Genomics ; 21(1): 143, 2020 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-32041546

RESUMEN

BACKGROUND: Successful mating of female mosquitoes typically occurs once, with the male sperm being stored in the female spermatheca for every subsequent oviposition event. The female spermatheca is responsible for the maintenance, nourishment, and protection of the male sperm against damage during storage. Aedes aegypti is a major vector of arboviruses, including Yellow Fever, Dengue, Chikungunya, and Zika. Vector control is difficult due to this mosquito high reproductive capacity. RESULTS: Following comparative RNA-seq analyses of spermathecae obtained from virgin and inseminated females, eight transcripts were selected based on their putative roles in sperm maintenance and survival, including energy metabolism, chitin components, transcriptional regulation, hormonal signaling, enzymatic activity, antimicrobial activity, and ionic homeostasis. In situ RNA hybridization confirmed tissue-specific expression of the eight transcripts. Following RNA interference (RNAi), observed outcomes varied between targeted transcripts, affecting mosquito survival, egg morphology, fecundity, and sperm motility within the spermathecae. CONCLUSIONS: This study identified spermatheca-specific transcripts associated with sperm storage in Ae. aegypti. Using RNAi we characterized the role of eight spermathecal transcripts on various aspects of female fecundity and offspring survival. RNAi-induced knockdown of transcript AeSigP-66,427, coding for a Na+/Ca2+ protein exchanger, specifically interfered with egg production and reduced sperm motility. Our results bring new insights into the molecular basis of sperm storage and identify potential targets for Ae. aegypti control.


Asunto(s)
Aedes/genética , Copulación , Genes de Insecto/fisiología , Inseminación , Mosquitos Vectores/genética , Motilidad Espermática , Animales , Femenino , Fertilidad/genética , Técnicas de Silenciamiento del Gen , Masculino , Interferencia de ARN , RNA-Seq , Espermatozoides/fisiología , Transcriptoma
6.
Malar J ; 19(1): 147, 2020 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-32268910

RESUMEN

BACKGROUND: A previous study reported that the malaria parasite Plasmodium falciparum enters an altered growth state upon extracellular withdrawal of the essential amino acid isoleucine. Parasites slowed transit through the cell cycle when deprived of isoleucine prior to the onset of S-phase. METHODS: This project was undertaken to study at higher resolution, how isoleucine withdrawal affects parasite growth. Parasites were followed at regular intervals across an extended isoleucine deprivation time course across the cell cycle using flow cytometry. RESULTS: These experiments revealed that isoleucine-deprived parasites never exit the cell cycle, but instead continuously grow at a markedly reduced pace. Moreover, slow growth occurs only if isoleucine is removed prior to the onset of schizogony. After S-phase commenced, the parasite is insensitive to isoleucine depletion and transits through the cell cycle at the normal pace. CONCLUSIONS: The markedly different response of the parasite to isoleucine withdrawal before or after the onset of DNA replication is reminiscent of the nutrient-dependent G1 cell cycle checkpoints described in other organisms.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , ADN Protozoario/fisiología , Eritrocitos/parasitología , Isoleucina/deficiencia , Plasmodium falciparum/crecimiento & desarrollo , Replicación del ADN/fisiología , Plasmodium falciparum/citología , Plasmodium falciparum/efectos de los fármacos
7.
Malar J ; 17(1): 367, 2018 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-30333029

RESUMEN

BACKGROUND: Using bacteria to express and deliver anti-parasite molecules in mosquitoes is among the list of genetic tools to control malaria. The introduction and spread of transgenic bacteria through wild adult mosquitoes is one of the major challenges of this strategy. In prospect of future field experiments, an open field study with blank (without bacteria) attractive sugar bait (ASB) was performed under the assumption that transgenic bacteria would be spread to all sugar fed mosquitoes. METHODS: Two types of ASB stations were developed, one with clay pots (CP) placed at mosquito resting sites and one with window entry traps (WET) placed inside inhabited houses. The ASB consisted in either glucose, honey or fruit cocktail solutions. In addition, mark-release-recapture (MRR) experiment of mosquitoes after feeding them with glucose was also conducted to check the proportion of the mosquito population that can be reached by the two ASB stations as well as its suitability to complement the ASB stations for disseminating bacteria. RESULTS: Overall, 88% of the mosquitoes were collected in the WET_ASB. The CP_ASB stations were much less attractive with the highest average of 82 ± 11 mosquitoes/day in the CP near the wood piles. The proportions of sugar fed mosquitoes upon ASB were low in both type of ASB stations, ~ 2% and ~ 14% in WET and CP, respectively. Honey solution was the most attractive solution compared to the glucose and the fruit cocktail solutions. The recapture rate in the MRR experiment was low: ~ 4.1% over 7 days. CONCLUSION: The WET_ASB looks promising to disseminate transgenic bacteria to endophilic West Africa Anopheles mosquito. However, this feeding station may not be fully effective and could be combined with the CP_ASB to also target outdoor resting mosquitoes. Overall, efforts are needed to improve the mosquito-feeding rates upon ASB.


Asunto(s)
Anopheles/fisiología , Control de Enfermedades Transmisibles/métodos , Conducta Alimentaria , Malaria/prevención & control , Control de Mosquitos/métodos , Animales , Burkina Faso , Carica , Citrullus , Control de Enfermedades Transmisibles/instrumentación , Femenino , Jugos de Frutas y Vegetales , Glucosa , Miel , Masculino , Estaciones del Año
8.
9.
Proc Natl Acad Sci U S A ; 112(26): E3412-20, 2015 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-26080400

RESUMEN

Plasmodium parasites must complete development in the mosquito vector for transmission to occur. The mosquito innate immune response is remarkably efficient in limiting parasite numbers. Previous work has identified a LPS-induced TNFα transcription factor (LITAF)-like transcription factor, LITAF-like 3 (LL3), which significantly influences parasite numbers. Here, we demonstrate that LL3 does not influence invasion of the mosquito midgut epithelium or ookinete-to-oocyst differentiation but mediates a late-phase immune response that decreases oocyst survival. LL3 expression in the midgut and hemocytes is activated by ookinete midgut invasion and is independent of the mosquito microbiota, suggesting that LL3 may be a component of a wound-healing response. LL3 silencing abrogates the ability of mosquito hemocytes to differentiate and respond to parasite infection, implicating hemocytes as critical modulators of the late-phase immune response.


Asunto(s)
Anopheles/inmunología , Anopheles/parasitología , Diferenciación Celular , Hemocitos/citología , Plasmodium/inmunología , Animales , Silenciador del Gen , Recuento de Huevos de Parásitos , Fagocitosis , Factores de Transcripción/genética , Factores de Transcripción/fisiología
10.
Exp Parasitol ; 181: 1-6, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28690071

RESUMEN

BACKGROUND: Plasmodium vivax infection remains a major public health problem, especially along the Thailand border regions. We examined the genetic diversity of this parasite by analyzing single-nucleotide polymorphisms (SNPs) of the P. vivax rhomboid-like protease 1 gene (Pvrom1) in parasites collected from western (Tak province, Thai-Myanmar border) and eastern (Chanthaburi province, Thai-Cambodia border) regions. METHODS: Data were collected by a cross-sectional survey, consisting of 47 and 45 P. vivax-infected filter paper-spotted blood samples from the western and eastern regions of Thailand, respectively during September 2013 to May 2014. Extracted DNA was examined for presence of P. vivax using Plasmodium species-specific nested PCR. Pvrom1 gene was PCR amplified, sequenced and the SNP diversity was analyzed using F-STAT, DnaSP, MEGA and LIAN programs. RESULTS: Comparison of sequences of the 92 Pvrom1 831-base open reading frames with that of a reference sequence (GenBank acc. no. XM001615211) revealed 17 samples with a total of 8 polymorphic sites, consisting of singleton (exon 3, nt 645) and parsimony informative (exon 1, nt 22 and 39; exon 3, nt 336, 537 and 656; and exon 4, nt 719 and 748) sites, which resulted in six different deduced Pvrom1 variants. Non-synonymous to synonymous substitutions ratio estimated by the DnaSP program was 1.65 indicating positive selection, but the Z-tests of selection showed no significant deviations from neutrality for Pvrom1 samples from western region of Thailand. In addition McDonald Kreitman test (MK) showed not significant, and Fst values are not different between the two regions and the regions combined. Interestingly, only Pvrom1 exon 2 was the most conserved sequences among the four exons. CONCLUSIONS: The relatively high degree of Pvrom1 polymorphism suggests that the protein is important for parasite survival in face of changes in both insect vector and human populations. These polymorphisms could serve as a sensitive marker for studying plasmodial genetic diversity. The significance of Pvrom1 conserved exon 2 sequence remains to be investigated.


Asunto(s)
Péptido Hidrolasas/genética , Plasmodium vivax/enzimología , Plasmodium vivax/genética , Polimorfismo de Nucleótido Simple , Proteínas Protozoarias/genética , Animales , Secuencia de Bases , Estudios Transversales , ADN Protozoario/química , ADN Protozoario/aislamiento & purificación , Exones/genética , Humanos , Insectos Vectores/parasitología , Desequilibrio de Ligamiento , Malaria Vivax/parasitología , Sistemas de Lectura Abierta , Péptido Hidrolasas/química , Reacción en Cadena de la Polimerasa , Proteínas Protozoarias/química , Especificidad de la Especie , Tailandia
11.
Proc Natl Acad Sci U S A ; 111(4): E492-500, 2014 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-24474798

RESUMEN

Plasmodium ookinete invasion of the mosquito midgut is a crucial step of the parasite life cycle but little is known about the molecular mechanisms involved. Previously, a phage display peptide library screen identified SM1, a peptide that binds to the mosquito midgut epithelium and inhibits ookinete invasion. SM1 was characterized as a mimotope of an ookinete surface enolase and SM1 presumably competes with enolase, the presumed ligand, for binding to a putative midgut receptor. Here we identify a mosquito midgut receptor that binds both SM1 and ookinete surface enolase, termed "enolase-binding protein" (EBP). Moreover, we determined that Plasmodium berghei parasites are heterogeneous for midgut invasion, as some parasite clones are strongly inhibited by SM1 whereas others are not. The SM1-sensitive parasites required the mosquito EBP receptor for midgut invasion whereas the SM1-resistant parasites invaded the mosquito midgut independently of EBP. These experiments provide evidence that Plasmodium ookinetes can invade the mosquito midgut by alternate pathways. Furthermore, another peptide from the original phage display screen, midgut peptide 2 (MP2), strongly inhibited midgut invasion by P. berghei (SM1-sensitive and SM1-resistant) and Plasmodium falciparum ookinetes, suggesting that MP2 binds to a separate, universal receptor for midgut invasion.


Asunto(s)
Abdomen/parasitología , Culicidae/parasitología , Plasmodium berghei/fisiología , Plasmodium falciparum/fisiología , Animales
12.
Cell Microbiol ; 17(11): 1594-604, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25944054

RESUMEN

Malaria remains one of the most devastating infectious diseases, killing up to a million people every year. Whereas much progress has been made in understanding the life cycle of the parasite in the human host and in the mosquito vector, significant gaps of knowledge remain. Fertilization of malaria parasites, a process that takes place in the lumen of the mosquito midgut, is poorly understood and the molecular interactions (receptor-ligand) required for Plasmodium fertilization remain elusive. By use of a phage display library, we identified FG1 (Female Gamete peptide 1), a peptide that binds specifically to the surface of female Plasmodium berghei gametes. Importantly, FG1 but not a scrambled version of the peptide, strongly reduces P. berghei oocyst formation by interfering with fertilization. In addition, FG1 also inhibits P. falciparum oocyst formation suggesting that the peptide binds to a molecule on the surface of the female gamete whose structure is conserved. Identification of the molecular interactions disrupted by the FG1 peptide may lead to the development of novel malaria transmission-blocking strategies.


Asunto(s)
División Celular , Plasmodium berghei/fisiología , Proteínas Protozoarias/metabolismo , Diferenciación Celular , Humanos , Proteínas Protozoarias/antagonistas & inhibidores
13.
Malar J ; 15(1): 394, 2016 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-27480269

RESUMEN

BACKGROUND: Malaria is transmitted when an infected mosquito delivers Plasmodium sporozoites into a vertebrate host. There are many species of Plasmodium and, in general, the infection is host-specific. For example, Plasmodium gallinaceum is an avian parasite, while Plasmodium berghei infects mice. These two parasites have been extensively used as experimental models of malaria transmission. Plasmodium falciparum and Plasmodium vivax are the most important agents of human malaria, a life-threatening disease of global importance. To complete their life cycle, Plasmodium parasites must traverse the mosquito midgut and form an oocyst that will divide continuously. Mature oocysts release thousands of sporozoites into the mosquito haemolymph that must reach the salivary gland to infect a new vertebrate host. The current understanding of the biology of oocyst formation and sporozoite release is mostly based on experimental infections with P. berghei, and the conclusions are generalized to other Plasmodium species that infect humans without further morphological analyses. RESULTS: Here, it is described the microanatomy of sporozoite escape from oocysts of four Plasmodium species: the two laboratory models, P. gallinaceum and P. berghei, and the two main species that cause malaria in humans, P. vivax and P. falciparum. It was found that sporozoites have species-specific mechanisms of escape from the oocyst. The two model species of Plasmodium had a common mechanism, in which the oocyst wall breaks down before sporozoites emerge. In contrast, P. vivax and P. falciparum sporozoites show a dynamic escape mechanism from the oocyst via polarized propulsion. CONCLUSIONS: This study demonstrated that Plasmodium species do not share a common mechanism of sporozoite escape, as previously thought, but show complex and species-specific mechanisms. In addition, the knowledge of this phenomenon in human Plasmodium can facilitate transmission-blocking studies and not those ones only based on the murine and avian models.


Asunto(s)
Oocistos/parasitología , Oocistos/ultraestructura , Plasmodium/fisiología , Plasmodium/ultraestructura , Esporozoítos/fisiología , Esporozoítos/ultraestructura , Animales , Aves , Femenino , Humanos , Estadios del Ciclo de Vida , Ratones , Microscopía Electrónica de Rastreo
14.
Eukaryot Cell ; 14(2): 128-39, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25446055

RESUMEN

The malaria parasite harbors a relict plastid called the apicoplast and its discovery opened a new avenue for drug discovery and development due to its unusual, nonmammalian metabolism. The apicoplast is essential during the asexual intraerythrocytic and hepatic stages of the parasite, and there is strong evidence supporting its essential metabolic role during the mosquito stages of the parasite. Supply of the isoprenoid building blocks isopentenyl diphosphate (IPP) and dimethylallyl diphosphate (DMAPP) is the essential metabolic function of the apicoplast during the asexual intraerythrocytic stages. However, the metabolic role of the apicoplast during gametocyte development, the malaria stages transmitted to the mosquito, remains unknown. In this study, we showed that production of IPP for isoprenoid biosynthesis is the essential metabolic function of the apicoplast during gametocytogenesis, by obtaining normal gametocytes lacking the apicoplast when supplemented with IPP. When IPP supplementation was removed early in gametocytogenesis, developmental defects were observed, supporting the essential role of isoprenoids for normal gametocytogenesis. Furthermore, mosquitoes infected with gametocytes lacking the apicoplast developed fewer and smaller oocysts that failed to produce sporozoites. This finding further supports the essential role of the apicoplast in establishing a successful infection in the mosquito vector. Our study supports isoprenoid biosynthesis as a valid drug target for development of malaria transmission-blocking inhibitors.


Asunto(s)
Apicoplastos/metabolismo , Hemiterpenos/biosíntesis , Estadios del Ciclo de Vida , Plasmodium falciparum/metabolismo , Animales , Gametogénesis , Compuestos Organofosforados , Plasmodium falciparum/crecimiento & desarrollo
15.
Proc Natl Acad Sci U S A ; 110(18): E1676-84, 2013 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-23572579

RESUMEN

Gametocytes are essential for Plasmodium transmission, but little is known about the mechanisms that lead to their formation. Using piggyBac transposon-mediated insertional mutagenesis, we screened for parasites that no longer form mature gametocytes, which led to the isolation of 29 clones (insertional gametocyte-deficient mutants) that fail to form mature gametocytes. Additional analysis revealed 16 genes putatively responsible for the loss of gametocytogenesis, none of which has been previously implicated in gametocytogenesis. Transcriptional profiling and detection of an early stage gametocyte antigen determined that a subset of these mutants arrests development at stage I or in early stage II gametocytes, likely representing genes involved in gametocyte maturation. The remaining mutants seem to arrest before formation of stage I gametocytes and may represent genes involved in commitment to the gametocyte lineage.


Asunto(s)
Elementos Transponibles de ADN/genética , Gametogénesis/genética , Genes Protozoarios/genética , Mutagénesis/genética , Plasmodium falciparum/genética , Animales , Prueba de Complementación Genética , Células Germinativas/metabolismo , Modelos Biológicos , Mutagénesis Insercional/genética , Mutación/genética , Parásitos/genética , Plásmidos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Antimicrob Agents Chemother ; 59(3): 1418-26, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25512427

RESUMEN

Reducing the transmission of the malarial parasite by Anopheles mosquitoes using drugs or vaccines remains a main focus in the efforts to control malaria. Iron chelators have been studied as potential antimalarial drugs due to their activities against different stages of the parasite. The iron chelator FBS0701 affects the development of Plasmodium falciparum early gametocytes and lowers blood-stage parasitemia. Here, we tested the effect of FBS0701 on stage V gametocyte infectivity for mosquitoes. The incubation of stage V gametocytes for up to 3 days with increasing concentrations of FBS0701 resulted in a significant dose-related reduction in mosquito infectivity, as measured by the numbers of oocysts per mosquito. The reduction in mosquito infectivity was due to the inhibition of male and female gametocyte activation. The preincubation of FBS0701 with ferric chloride restored gametocyte infectivity, showing that the inhibitory effect of FBS0701 was quenched by iron. Deferoxamine, another iron chelator, also reduced gametocyte infectivity but to a lesser extent. Finally, the simultaneous administration of drug and gametocytes to mosquitoes without previous incubation did not significantly reduce the numbers of oocysts. These results show the importance of gametocyte iron metabolism as a potential target for new transmission-blocking strategies.


Asunto(s)
Antimaláricos/farmacología , Éteres de Etila/farmacología , Quelantes del Hierro/farmacología , Plasmodium falciparum/efectos de los fármacos , Tiazoles/farmacología , Animales , Deferoxamina/farmacología , Femenino , Masculino
17.
Proc Natl Acad Sci U S A ; 109(31): 12734-9, 2012 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-22802646

RESUMEN

The most vulnerable stages of Plasmodium development occur in the lumen of the mosquito midgut, a compartment shared with symbiotic bacteria. Here, we describe a strategy that uses symbiotic bacteria to deliver antimalaria effector molecules to the midgut lumen, thus rendering host mosquitoes refractory to malaria infection. The Escherichia coli hemolysin A secretion system was used to promote the secretion of a variety of anti-Plasmodium effector proteins by Pantoea agglomerans, a common mosquito symbiotic bacterium. These engineered P. agglomerans strains inhibited development of the human malaria parasite Plasmodium falciparum and rodent malaria parasite Plasmodium berghei by up to 98%. Significantly, the proportion of mosquitoes carrying parasites (prevalence) decreased by up to 84% for two of the effector molecules, scorpine, a potent antiplasmodial peptide and (EPIP)(4), four copies of Plasmodium enolase-plasminogen interaction peptide that prevents plasminogen binding to the ookinete surface. We demonstrate the use of an engineered symbiotic bacterium to interfere with the development of P. falciparum in the mosquito. These findings provide the foundation for the use of genetically modified symbiotic bacteria as a powerful tool to combat malaria.


Asunto(s)
Anopheles , Antimaláricos/metabolismo , Proteínas de Escherichia coli/biosíntesis , Proteínas Hemolisinas/biosíntesis , Insectos Vectores , Malaria Falciparum/prevención & control , Pantoea/metabolismo , Plasmodium berghei , Plasmodium falciparum , Animales , Anopheles/metabolismo , Anopheles/microbiología , Anopheles/parasitología , Sistemas de Secreción Bacterianos/genética , Proteínas de Escherichia coli/genética , Proteínas Hemolisinas/genética , Insectos Vectores/inmunología , Insectos Vectores/parasitología , Malaria Falciparum/metabolismo , Pantoea/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Simbiosis
18.
PLoS Pathog ; 8(10): e1002965, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23093936

RESUMEN

The mosquito is the obligate vector for malaria transmission. To complete its development within the mosquito, the malaria parasite Plasmodium must overcome the protective action of the mosquito innate immune system. Here we report on the involvement of the Anopheles gambiae orthologue of a conserved component of the vertebrate immune system, LPS-induced TNFα transcription factor (LITAF), and its role in mosquito anti-Plasmodium immunity. An. gambiae LITAF-like 3 (LL3) expression is up-regulated in response to midgut invasion by both rodent and human malaria parasites. Silencing of LL3 expression greatly increases parasite survival, indicating that LL3 is part of an anti-Plasmodium defense mechanism. Electrophoretic mobility shift assays identified specific LL3 DNA-binding motifs within the promoter of SRPN6, a gene that also mediates mosquito defense against Plasmodium. Further experiments indicated that these motifs play a direct role in LL3 regulation of SRPN6 expression. We conclude that LL3 is a transcription factor capable of modulating SRPN6 expression as part of the mosquito anti-Plasmodium immune response.


Asunto(s)
Anopheles/inmunología , Interacciones Huésped-Parásitos , Proteínas de Insectos/metabolismo , Insectos Vectores/inmunología , Plasmodium/inmunología , Factores de Transcripción/metabolismo , Animales , Anopheles/genética , Anopheles/parasitología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Insectos/genética , Insectos Vectores/parasitología , Malaria/transmisión , Plasmodium/genética , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño , Factores de Transcripción/genética
19.
Proc Natl Acad Sci U S A ; 108(41): 17153-8, 2011 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-21949403

RESUMEN

Ookinete invasion of the mosquito midgut is an essential step for the development of the malaria parasite in the mosquito. Invasion involves recognition between a presumed mosquito midgut receptor and an ookinete ligand. Here, we show that enolase lines the ookinete surface. An antienolase antibody inhibits oocyst development of both Plasmodium berghei and Plasmodium falciparum, suggesting that enolase may act as an invasion ligand. Importantly, we demonstrate that surface enolase captures plasminogen from the mammalian blood meal via its lysine motif (DKSLVK) and that this interaction is essential for midgut invasion, because plasminogen depletion leads to a strong inhibition of oocyst formation. Although addition of recombinant WT plasminogen to depleted serum rescues oocyst formation, recombinant inactive plasminogen does not, thus emphasizing the importance of plasmin proteolytic activity for ookinete invasion. The results support the hypothesis that enolase on the surface of Plasmodium ookinetes plays a dual role in midgut invasion: by acting as a ligand that interacts with the midgut epithelium and, further, by capturing plasminogen, whose conversion to active plasmin promotes the invasion process.


Asunto(s)
Anopheles/parasitología , Plasminógeno/fisiología , Plasmodium berghei/fisiología , Plasmodium berghei/patogenicidad , Plasmodium falciparum/fisiología , Plasmodium falciparum/patogenicidad , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Sistema Digestivo/parasitología , Humanos , Insectos Vectores/parasitología , Modelos Biológicos , Oocistos/fisiología , Fosfopiruvato Hidratasa/genética , Fosfopiruvato Hidratasa/fisiología , Plasminógeno/química , Plasminógeno/genética , Plasmodium berghei/crecimiento & desarrollo , Plasmodium falciparum/genética , Plasmodium falciparum/crecimiento & desarrollo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
20.
Mem Inst Oswaldo Cruz ; 109(5): 644-61, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25185005

RESUMEN

Nearly one million people are killed every year by the malaria parasite Plasmodium. Although the disease-causing forms of the parasite exist only in the human blood, mosquitoes of the genus Anopheles are the obligate vector for transmission. Here, we review the parasite life cycle in the vector and highlight the human and mosquito contributions that limit malaria parasite development in the mosquito host. We address parasite killing in its mosquito host and bottlenecks in parasite numbers that might guide intervention strategies to prevent transmission.


Asunto(s)
Anopheles/parasitología , Insectos Vectores/parasitología , Estadios del Ciclo de Vida/fisiología , Plasmodium/crecimiento & desarrollo , Animales , Anopheles/clasificación , Interacciones Huésped-Parásitos/fisiología , Humanos , Insectos Vectores/clasificación , Malaria/transmisión , Plasmodium/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA