Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(44): e2304339120, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37883438

RESUMEN

Malaria remains a devastating disease and, with current measures failing to control its transmission, there is a need for novel interventions. A family of proteins that have long been pursued as potential intervention targets are aquaporins, which are channels facilitating the movement of water and other solutes across membranes. We identify an aquaporin in malaria parasites and demonstrate that it is important for completion of Plasmodium development in the mosquito vector. Disruption of AQP2 in the human parasite Plasmodium falciparum and the rodent parasite Plasmodium berghei blocks sporozoite production inside oocysts established on mosquito midguts, greatly limiting parasite infection of salivary glands and transmission to a new host. In vivo epitope tagging of AQP2 in P. berghei, combined with immunofluorescence assays, reveals that the protein is localized in vesicle-like organelles found in the cytoplasm of gametocytes, ookinetes, and sporozoites. The number of these organelles varies between individual parasites and lifecycle stages suggesting that they are likely part of a dynamic endomembrane system. Phylogenetic analysis confirms that AQP2 is unique to malaria and closely related parasites and most closely resembles intracellular aquaporins. Structure prediction analyses identify several unusual features, including a large accessory extracellular loop and an arginine-to-phenylalanine substitution in the selectivity filter principally determining pore function, a unique feature among known aquaporins. This in conjunction with the importance of AQP2 for malaria transmission suggests that AQP2 may be a fruitful target of antimalarial interventions.


Asunto(s)
Acuaporina 2 , Mosquitos Vectores , Proteínas Protozoarias , Animales , Malaria , Mosquitos Vectores/parasitología , Filogenia , Plasmodium berghei/metabolismo , Proteínas Protozoarias/metabolismo , Esporozoítos/metabolismo
2.
PLoS Pathog ; 17(5): e1009486, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34015060

RESUMEN

Vitellogenesis and oocyte maturation require anautogenous female Anopheles mosquitoes to obtain a bloodmeal from a vertebrate host. The bloodmeal is rich in proteins that are readily broken down into amino acids in the midgut lumen and absorbed by the midgut epithelial cells where they are converted into lipids and then transported to other tissues including ovaries. The stearoyl-CoA desaturase (SCD) plays a pivotal role in this process by converting saturated (SFAs) to unsaturated (UFAs) fatty acids; the latter being essential for maintaining cell membrane fluidity amongst other housekeeping functions. Here, we report the functional and phenotypic characterization of SCD1 in the malaria vector mosquito Anopheles coluzzii. We show that RNA interference (RNAi) silencing of SCD1 and administration of sterculic acid (SA), a small molecule inhibitor of SCD1, significantly impact on the survival and reproduction of female mosquitoes following blood feeding. Microscopic observations reveal that the mosquito thorax is quickly filled with blood, a phenomenon likely caused by the collapse of midgut epithelial cell membranes, and that epithelial cells are depleted of lipid droplets and oocytes fail to mature. Transcriptional profiling shows that genes involved in protein, lipid and carbohydrate metabolism and immunity-related genes are the most affected by SCD1 knock down (KD) in blood-fed mosquitoes. Metabolic profiling reveals that these mosquitoes exhibit increased amounts of saturated fatty acids and TCA cycle intermediates, highlighting the biochemical framework by which the SCD1 KD phenotype manifests as a result of a detrimental metabolic syndrome. Accumulation of SFAs is also the likely cause of the potent immune response observed in the absence of infection, which resembles an auto-inflammatory condition. These data provide insights into mosquito bloodmeal metabolism and lipid homeostasis and could inform efforts to develop novel interventions against mosquito-borne diseases.


Asunto(s)
Alimentación Animal/análisis , Anopheles/crecimiento & desarrollo , Conducta Alimentaria , Mosquitos Vectores/fisiología , Reproducción , Estearoil-CoA Desaturasa/metabolismo , Animales , Anopheles/enzimología , Anopheles/inmunología , Femenino , Perfilación de la Expresión Génica , Mosquitos Vectores/parasitología , Estearoil-CoA Desaturasa/genética
3.
Proc Natl Acad Sci U S A ; 117(13): 7363-7373, 2020 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-32165544

RESUMEN

After being ingested by a female Anopheles mosquito during a bloodmeal on an infected host, and before they can reach the mosquito salivary glands to be transmitted to a new host, Plasmodium parasites must establish an infection of the mosquito midgut in the form of oocysts. To achieve this, they must first survive a series of robust innate immune responses that take place prior to, during, and immediately after ookinete traversal of the midgut epithelium. Understanding how parasites may evade these responses could highlight new ways to block malaria transmission. We show that an ookinete and sporozoite surface protein designated as PIMMS43 (Plasmodium Infection of the Mosquito Midgut Screen 43) is required for parasite evasion of the Anopheles coluzzii complement-like response. Disruption of PIMMS43 in the rodent malaria parasite Plasmodium berghei triggers robust complement activation and ookinete elimination upon mosquito midgut traversal. Silencing components of the complement-like system through RNAi largely restores ookinete-to-oocyst transition but oocysts remain small in size and produce a very small number of sporozoites that additionally are not infectious, indicating that PIMMS43 is also essential for sporogonic development in the oocyst. Antibodies that bind PIMMS43 interfere with parasite immune evasion when ingested with the infectious blood meal and significantly reduce the prevalence and intensity of infection. PIMMS43 genetic structure across African Plasmodium falciparum populations indicates allelic adaptation to sympatric vector populations. These data add to our understanding of mosquito-parasite interactions and identify PIMMS43 as a target of malaria transmission blocking.


Asunto(s)
Anopheles/inmunología , Mosquitos Vectores/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Animales , Anopheles/metabolismo , Anopheles/parasitología , Femenino , Interacciones Huésped-Parásitos/inmunología , Humanos , Evasión Inmune , Malaria Falciparum/parasitología , Malaria Falciparum/transmisión , Mosquitos Vectores/metabolismo , Mosquitos Vectores/parasitología , Oocistos/inmunología , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Esporozoítos/inmunología
4.
Infect Immun ; 85(8)2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28559405

RESUMEN

Mosquito midgut stages of the malaria parasite present an attractive biological system to study host-parasite interactions and develop interventions to block disease transmission. Mosquito infection ensues upon oocyst development that follows ookinete invasion and traversal of the mosquito midgut epithelium. Here, we report the characterization of PIMMS2 (Plasmodium invasion of mosquito midgut screen candidate 2), a Plasmodium berghei protein with structural similarities to subtilisin-like proteins. PIMMS2 orthologs are present in the genomes of all plasmodia and are mapped between the subtilisin-encoding genes SUB1 and SUB3 P. berghei PIMMS2 is specifically expressed in zygotes and ookinetes and is localized on the ookinete surface. Loss of PIMMS2 function through gene disruption by homologous recombination leads to normal development of motile ookinetes that exhibit a severely impaired capacity to traverse the mosquito midgut and transform to oocysts. Genetic complementation of the disrupted locus with a mutated PIMMS2 allele reveals that amino acid residues corresponding to the putative subtilisin-like catalytic triad are important but not essential for protein function. Our data demonstrate that PIMMS2 is a novel ookinete-specific protein that promotes parasite traversal of the mosquito midgut epithelium and establishment of mosquito infection.

5.
Cell Microbiol ; 17(8): 1230-40, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25728487

RESUMEN

The malaria parasite develops sexually in the mosquito midgut upon entry with the ingested blood meal before it can invade the midgut epithelium and embark on sporogony. Recent data have identified a number of distinct transcriptional programmes operating during this critical phase of the parasite life cycle. We aimed at characterizing the parental contribution to these transcriptional programmes and establish the genetic framework that would guide further studies of Plasmodium zygotic development and ookinete-to-oocyst transition. To achieve this we used in vitro and in vivo cross-fertilization experiments of various parasite lines expressing fluorescent reporters under the control of constitutive and stage-specific promoters. The results revealed that the zygote/ookinete stage exhibits a maternal phenotype with respect to constitutively expressed reporters, which is derived from either maternal mRNA inheritance or transcription of the maternal allele. The respective paternal alleles are silenced in the zygote/ookinete but reactivated after midgut invasion and transformation to oocyst. Transcripts specifically produced in the zygote/ookinete are synthesized de novo by both parental alleles. These findings highlight a putative role of epigenetic regulation of Plasmodium zygotic development and add substantially to the emerging picture of the molecular mechanisms regulating this important stage of malaria transmission.


Asunto(s)
ADN Protozoario/genética , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Genes Protozoarios , Oocistos , Plasmodium berghei/genética , Activación Transcripcional , Fusión Artificial Génica , Perfilación de la Expresión Génica , Genes Reporteros , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/genética , Regiones Promotoras Genéticas , Transcripción Genética
6.
Cell Microbiol ; 17(2): 254-68, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25225164

RESUMEN

The passage through the mosquito is a major bottleneck for malaria parasite populations and a target of interventions aiming to block disease transmission. Here, we used DNA microarrays to profile the developmental transcriptomes of the rodent malaria parasite Plasmodium berghei in vivo, in the midgut of Anopheles gambiae mosquitoes, from parasite stages in the midgut blood bolus to sporulating oocysts on the basal gut wall. Data analysis identified several distinct transcriptional programmes encompassing genes putatively involved in developmental processes or in interactions with the mosquito. At least two of these programmes are associated with the ookinete development that is linked to mosquito midgut invasion and establishment of infection. Targeted disruption by homologous recombination of two of these genes resulted in mutant parasites exhibiting notable infection phenotypes. GAMER encodes a short polypeptide with granular localization in the gametocyte cytoplasm and shows a highly penetrant loss-of-function phenotype manifested as greatly reduced ookinete numbers, linked to impaired male gamete release. HADO encodes a putative magnesium phosphatase with distinctive cortical localization along the concave ookinete periphery. Disruption of HADO compromises ookinete development leading to significant reduction of oocyst numbers. Our data provide important insights into the molecular framework underpinning Plasmodium development in the mosquito and identifies two genes with important functions at initial stages of parasite development in the mosquito midgut.


Asunto(s)
Anopheles/parasitología , Perfilación de la Expresión Génica , Plasmodium berghei/crecimiento & desarrollo , Animales , Tracto Gastrointestinal/parasitología , Malaria/transmisión , Análisis de Secuencia por Matrices de Oligonucleótidos , Plasmodium berghei/genética , Plasmodium berghei/aislamiento & purificación
7.
Cell Host Microbe ; 31(9): 1539-1551.e6, 2023 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-37708854

RESUMEN

Malaria remains one of the most devastating infectious diseases. Reverse genetic screens offer a powerful approach to identify genes and molecular processes governing malaria parasite biology. However, the complex regulation of gene expression and genotype-phenotype associations in the mosquito vector, along with sexual reproduction, have hindered the development of screens in this critical part of the parasite life cycle. To address this, we developed a genetic approach in the rodent parasite Plasmodium berghei that, in combination with barcode sequencing, circumvents the fertilization roadblock and enables screening for gametocyte-expressed genes required for parasite infection of the mosquito Anopheles coluzzii. Our results confirm previous findings, validating our approach for scaling up, and identify genes necessary for mosquito midgut infection, oocyst development, and salivary gland infection. These findings can aid efforts to study malaria transmission biology and to develop interventions for controlling disease transmission.


Asunto(s)
Anopheles , Esporozoítos , Animales , Esporozoítos/genética , Mosquitos Vectores/genética , Plasmodium berghei/genética , Anopheles/genética
8.
Infect Immun ; 79(11): 4708-15, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21844236

RESUMEN

Malaria remains a devastating disease despite efforts at control and prevention. Extensive studies using mostly rodent infection models reveal that successful Plasmodium parasite transmission by the African mosquito vector Anopheles gambiae depends on finely tuned vector-parasite interactions. Here we investigate the transcriptional response of A. gambiae to geographically related Plasmodium falciparum populations at various infection intensities and different infection stages. These responses are compared with those of mosquitoes infected with the rodent parasite Plasmodium berghei. We demonstrate that mosquito responses are largely dependent on the intensity of infection. A major transcriptional suppression of genes involved in the regulation of midgut homeostasis is detected in low-intensity P. falciparum infections, the most common type of infection in Africa. Importantly, genes transcriptionally induced during these infections tend to be phylogenetically unique to A. gambiae. These data suggest that coadaptation between vectors and parasites may act to minimize the impact of infection on mosquito fitness by selectively suppressing specific functional classes of genes. RNA interference (RNAi)-mediated gene silencing provides initial evidence for important roles of the mosquito G protein-coupled receptors (GPCRs) in controlling infection intensity-dependent antiparasitic responses.


Asunto(s)
Anopheles/inmunología , Anopheles/parasitología , Insectos Vectores/inmunología , Insectos Vectores/parasitología , Plasmodium falciparum/fisiología , Animales , Anopheles/genética , Anopheles/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Interacciones Huésped-Parásitos , Humanos , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Insectos Vectores/genética , Insectos Vectores/metabolismo , Ratones , Filogenia , Plasmodium berghei/fisiología
9.
PLoS Pathog ; 5(8): e1000539, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19662167

RESUMEN

Malaria parasites must undergo sexual and sporogonic development in mosquitoes before they can infect their vertebrate hosts. We report the discovery and characterization of MISFIT, the first protein with paternal effect on the development of the rodent malaria parasite Plasmodium berghei in Anopheles mosquitoes. MISFIT is expressed in male gametocytes and localizes to the nuclei of male gametocytes, zygotes and ookinetes. Gene disruption results in mutant ookinetes with reduced genome content, microneme defects and altered transcriptional profiles of putative cell cycle regulators, which yet successfully invade the mosquito midgut. However, developmental arrest ensues during the ookinete transformation to oocysts leading to malaria transmission blockade. Genetic crosses between misfit mutant parasites and parasites that are either male or female gamete deficient reveal a strict requirement for a male misfit allele. MISFIT belongs to the family of formin-like proteins, which are known regulators of the dynamic remodeling of actin and microtubule networks. Our data identify the ookinete-to-oocyst transition as a critical cell cycle checkpoint in Plasmodium development and lead us to hypothesize that MISFIT may be a regulator of cell cycle progression. This study offers a new perspective for understanding the male contribution to malaria parasite development in the mosquito vector.


Asunto(s)
Culicidae/parasitología , Insectos Vectores/parasitología , Plasmodium berghei/crecimiento & desarrollo , Proteínas Protozoarias/genética , Secuencia de Aminoácidos , Animales , Southern Blotting , Femenino , Técnica del Anticuerpo Fluorescente , Expresión Génica , Regulación de la Expresión Génica , Genes Protozoarios/genética , Malaria/transmisión , Masculino , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Filogenia , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Proteínas Protozoarias/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Nucleic Acids Res ; 37(11): 3788-98, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19380379

RESUMEN

Techniques for targeted genetic disruption in Plasmodium, the causative agent of malaria, are currently intractable for those genes that are essential for blood stage development. The ability to use RNA interference (RNAi) to silence gene expression would provide a powerful means to gain valuable insight into the pathogenic blood stages but its functionality in Plasmodium remains controversial. Here we have used various RNA-based gene silencing approaches to test the utility of RNAi in malaria parasites and have undertaken an extensive comparative genomics search using profile hidden Markov models to clarify whether RNAi machinery exists in malaria. These investigative approaches revealed that Plasmodium lacks the enzymology required for RNAi-based ablation of gene expression and indeed no experimental evidence for RNAi was observed. In its absence, the most likely explanations for previously reported RNAi-mediated knockdown are either the general toxicity of introduced RNA (with global down-regulation of gene expression) or a specific antisense effect mechanistically distinct from RNAi, which will need systematic analysis if it is to be of use as a molecular genetic tool for malaria parasites.


Asunto(s)
Genoma de Protozoos , Plasmodium berghei/genética , Plasmodium falciparum/genética , Interferencia de ARN , Animales , Genes Protozoarios , Genómica , Plasmodium berghei/metabolismo , Plasmodium falciparum/enzimología , Plasmodium falciparum/metabolismo , ARN sin Sentido/metabolismo , ARN Bicatenario/metabolismo , ARN Interferente Pequeño/metabolismo , Ribonucleasa III/genética
11.
Front Cell Infect Microbiol ; 11: 634273, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33791240

RESUMEN

Plasmodium falciparum malaria remains a major cause of global morbidity and mortality, mainly in sub-Saharan Africa. The numbers of new malaria cases and deaths have been stable in the last years despite intense efforts for disease elimination, highlighting the need for new approaches to stop disease transmission. Further understanding of the parasite transmission biology could provide a framework for the development of such approaches. We phenotypically and functionally characterized three novel genes, PIMMS01, PIMMS57, and PIMMS22, using targeted disruption of their orthologs in the rodent parasite Plasmodium berghei. PIMMS01 and PIMMS57 are specifically and highly expressed in ookinetes, while PIMMS22 transcription starts already in gametocytes and peaks in sporozoites. All three genes show strong phenotypes associated with the ookinete to oocyst transition, as their disruption leads to very low numbers of oocysts and complete abolishment of transmission. PIMMS22 has a secondary essential function in the oocyst. Our results enrich the molecular understanding of the parasite-vector interactions and identify PIMMS01, PIMMS57, and PIMMS22 as new targets of transmission blocking interventions.


Asunto(s)
Malaria Falciparum , Malaria , Animales , Oocistos , Plasmodium berghei , Esporozoítos
12.
PLoS Pathog ; 4(5): e1000069, 2008 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-18483558

RESUMEN

In much of sub-Saharan Africa, the mosquito Anopheles gambiae is the main vector of the major human malaria parasite, Plasmodium falciparum. Convenient laboratory studies have identified mosquito genes that affect positively or negatively the developmental cycle of the model rodent parasite, P. berghei. Here, we use transcription profiling and reverse genetics to explore whether five disparate mosquito gene regulators of P. berghei development are also pertinent to A. gambiae/P. falciparum interactions in semi-natural conditions, using field isolates of this parasite and geographically related mosquitoes. We detected broadly similar albeit not identical transcriptional responses of these genes to the two parasite species. Gene silencing established that two genes affect similarly both parasites: infections are hindered by the intracellular local activator of actin cytoskeleton dynamics, WASP, but promoted by the hemolymph lipid transporter, ApoII/I. Since P. berghei is not a natural parasite of A. gambiae, these data suggest that the effects of these genes have not been drastically altered by constant interaction and co-evolution of A. gambiae and P. falciparum; this conclusion allowed us to investigate further the mode of action of these two genes in the laboratory model system using a suite of genetic tools and infection assays. We showed that both genes act at the level of midgut invasion during the parasite's developmental transition from ookinete to oocyst. ApoII/I also affects the early stages of oocyst development. These are the first mosquito genes whose significant effects on P. falciparum field isolates have been established by direct experimentation. Importantly, they validate for semi-field human malaria transmission the concept of parasite antagonists and agonists.


Asunto(s)
Anopheles/parasitología , Vectores de Enfermedades , Malaria Falciparum/parasitología , Plasmodium berghei/fisiología , Plasmodium falciparum/fisiología , Animales , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Apolipoproteína A-II/genética , Apolipoproteína A-II/metabolismo , Niño , Preescolar , ADN Protozoario/análisis , Sistema Digestivo/parasitología , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Silenciador del Gen , Predisposición Genética a la Enfermedad , Interacciones Huésped-Parásitos , Humanos , Malaria Falciparum/sangre , Malaria Falciparum/transmisión , Oocistos/fisiología , Plasmodium berghei/patogenicidad , Plasmodium falciparum/patogenicidad , Polimorfismo de Nucleótido Simple , Transcripción Genética , Proteína del Síndrome de Wiskott-Aldrich/genética , Proteína del Síndrome de Wiskott-Aldrich/metabolismo
13.
Curr Opin Genet Dev ; 16(4): 384-91, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16793259

RESUMEN

Migration of the protozoan parasite Plasmodium through the mosquito is a complex and delicate process, the outcome of which determines the success of malaria transmission. The mosquito is not simply the vector of Plasmodium but, in terms of the life cycle, its definitive host: there, the parasite undergoes its sexual development, which results in colonization of the mosquito salivary glands. Two of the parasite's developmental stages in the mosquito, the ookinete and the sporozoite, are invasive and depend on gliding motility to access, penetrate and traverse their host cells. Recent advances in the field have included the identification of numerous Plasmodium molecules that are essential for parasite migration in the mosquito vector.


Asunto(s)
Culicidae/parasitología , Plasmodium/crecimiento & desarrollo , Animales , Movimiento , Oocistos/crecimiento & desarrollo , Plasmodium/fisiología , Glándulas Salivales/parasitología , Esporozoítos/crecimiento & desarrollo
14.
Sci Rep ; 10(1): 6354, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32286373

RESUMEN

The malaria parasite replicates asexually in the red blood cells of its vertebrate host employing epigenetic mechanisms to regulate gene expression in response to changes in its environment. We used chromatin immunoprecipitation followed by sequencing in conjunction with RNA sequencing to create an epigenomic and transcriptomic map of the developmental transition from asexual blood stages to male and female gametocytes and to ookinetes in the rodent malaria parasite Plasmodium berghei. Across the developmental stages examined, heterochromatin protein 1 associates with variantly expressed gene families localised at subtelomeric regions and variant gene expression based on heterochromatic silencing is observed only in some genes. Conversely, the euchromatin mark histone 3 lysine 9 acetylation (H3K9ac) is abundant in non-heterochromatic regions across all developmental stages. H3K9ac presents a distinct pattern of enrichment around the start codon of ribosomal protein genes in all stages but male gametocytes. Additionally, H3K9ac occupancy positively correlates with transcript abundance in all stages but female gametocytes suggesting that transcription in this stage is independent of H3K9ac levels. This finding together with known mRNA repression in female gametocytes suggests a multilayered mechanism operating in female gametocytes in preparation for fertilization and zygote development, coinciding with parasite transition from host to vector.


Asunto(s)
Epigénesis Genética/genética , Código de Histonas/genética , Malaria Falciparum/genética , Plasmodium falciparum/genética , Acetilación , Animales , Cromatina/genética , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/genética , Fertilización/genética , Regulación de la Expresión Génica/genética , Células Germinativas/crecimiento & desarrollo , Células Germinativas/metabolismo , Humanos , Malaria Falciparum/parasitología , Malaria Falciparum/patología , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/patogenicidad , Procesamiento Proteico-Postraduccional/genética , Análisis de Secuencia de ARN , Cigoto/crecimiento & desarrollo , Cigoto/metabolismo
15.
Curr Biol ; 15(13): 1185-95, 2005 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-16005290

RESUMEN

BACKGROUND: The malaria parasite Plasmodium must complete a complex developmental life cycle within Anopheles mosquitoes before it can be transmitted into the human host. One day after mosquito infection, motile ookinetes traverse the midgut epithelium and, after exiting to its basal site facing the hemolymph, develop into oocysts. Previously, we have identified hemolymph factors that can antagonize or promote parasite development. RESULTS: We profiled on a genomic scale the transcriptional responses of the A. gambiae midgut to P. berghei and showed that more than 7% of the assessed mosquito transcriptome is differentially regulated during invasion. The profiles suggested that actin- and microtubule-cytoskeleton remodeling is a major response of the epithelium to ookinete penetration. Other responses encompass components of innate immunity, extracellular-matrix remodeling, and apoptosis. RNAi-dependent gene silencing identified both parasite antagonists and agonists among regulators of actin dynamics and revealed that actin polymerization is inhibitory to the invading parasite. Combined transcriptional and reverse-genetic analysis further identified an unexpected dual role of the lipid-trafficking machinery of the hemolymph for both parasite and mosquito-egg development. CONCLUSIONS: We conclude that the determinants of malaria-parasite development in Anopheles include components not only of systemic humoral immunity but also of intracellular, local epithelial reactions. These results provide novel mechanistic insights for understanding malaria transmission in the mosquito vector.


Asunto(s)
Anopheles/genética , Anopheles/parasitología , Sistema Digestivo/parasitología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Plasmodium berghei , Actinas/metabolismo , Animales , Anopheles/metabolismo , Apoptosis/genética , Análisis por Conglomerados , Epitelio/metabolismo , Epitelio/parasitología , Genómica/métodos , Inmunidad Innata/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Curr Opin Microbiol ; 8(4): 415-21, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15996894

RESUMEN

The malaria parasite, Plasmodium, requires sexual development in the mosquito before it can be transmitted to the vertebrate host. Mosquito genes are able to substantially modulate this process, which can result in major decreases in parasite numbers. Even in susceptible mosquitoes, haemolymph proteins implicated in systemic immune reactions, together with local epithelial responses, cause lysis of more than 80% of the ookinetes that cross the mosquito midgut. In a refractory mosquito strain, immune responses lead to melanisation of virtually all parasites. Conversely, certain mosquito genes have an opposite effect: they are used by the parasite to evade defence reactions. Detailed understanding of the interplay between positive and negative regulators of parasite development could lead to the generation of novel approaches for malaria control through the vector.


Asunto(s)
Anopheles/parasitología , Regulación del Desarrollo de la Expresión Génica , Insectos Vectores/parasitología , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/fisiología , Animales , Interacciones Huésped-Parásitos , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo
17.
Sci Rep ; 7(1): 6026, 2017 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-28729672

RESUMEN

Malaria is a mosquito-borne disease affecting millions of people every year. The rodent parasite Plasmodium berghei has served as a model for human malaria transmission studies and played a pivotal role in dissecting the mosquito immune response against infection. The 6-cysteine protein P47, known to be important for P. berghei female gamete fertility, is shown to serve a different function in Plasmodium falciparum, protecting ookinetes from the mosquito immune response. Here, we investigate the function of P. berghei P47 in Anopheles gambiae mosquito infections. We show that P47 is expressed on the surface of both female gametocytes and ookinetes where it serves distinct functions in promoting gametocyte-to-ookinete development and protecting ookinetes from the mosquito complement-like response, respectively. The latter function is essential, as ookinetes lacking P47 are targeted for killing while traversing the mosquito midgut cells and eliminated upon exposure to hemolymph proteins of the complement-like system. Silencing key factors of the complement-like system restores oocyst development and disease transmission to rodent hosts. Our data establish a dual role of P. berghei P47 in vivo and reinforce the use of this parasite to study the impact of the mosquito immune response on human malaria transmission.


Asunto(s)
Anopheles/inmunología , Anopheles/parasitología , Interacciones Huésped-Parásitos/inmunología , Plasmodium berghei/fisiología , Proteínas Protozoarias/metabolismo , Animales , Proteínas del Sistema Complemento/inmunología , Femenino , Expresión Génica , Silenciador del Gen , Malaria/parasitología , Malaria/transmisión , Oocistos , Proteínas Protozoarias/genética , Eliminación de Secuencia
18.
Pathog Glob Health ; 107(8): 480-92, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24428832

RESUMEN

Successful completion of the Plasmodium lifecycle in the mosquito vector is critical for malaria transmission. It has been documented that the fate of Plasmodium in the mosquito ultimately depends on a fine interplay of molecular mosquito factors that act as parasite agonists and antagonists. Here we investigate whether the cellular responses of the invaded midgut epithelium can also determine the parasite fate and development. We show that the parasite hood, an actin-rich structure formed around the ookinete as it exits the epithelium, is a local epithelial defence reaction observed around 60% of invading parasites. The hood co-localizes with WASP, a promoter of actin filament nucleation, suggesting that it is an active reaction of the invaded cell against invading parasites. Importantly, depletion of WASP by RNAi leads to a significant reduction in hood formation, which is consistent with the previously documented role of this gene as a potent parasite antagonist. Indeed, in mosquitoes that are either genetically selected or manipulated by RNAi to be refractory to Plasmodium, most dead parasites exhibit an actin hood. In these mosquitoes, invading ookinetes are killed by lysis or melanization while exiting the midgut epithelium. Silencing WASP in these mosquitoes inhibits the formation of the hood and allows many parasites to develop to oocysts. These data in conjunction with fine microscopic observations suggest that the presence of the hood is linked to ookinete killing through lysis.


Asunto(s)
Actinas/metabolismo , Culicidae/inmunología , Culicidae/parasitología , Células Epiteliales/inmunología , Células Epiteliales/parasitología , Interacciones Huésped-Patógeno , Plasmodium/crecimiento & desarrollo , Animales , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/parasitología , Proteína del Síndrome de Wiskott-Aldrich/metabolismo
19.
Science ; 316(5832): 1738-43, 2007 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-17588928

RESUMEN

Mosquitoes are vectors of parasitic and viral diseases of immense importance for public health. The acquisition of the genome sequence of the yellow fever and Dengue vector, Aedes aegypti (Aa), has enabled a comparative phylogenomic analysis of the insect immune repertoire: in Aa, the malaria vector Anopheles gambiae (Ag), and the fruit fly Drosophila melanogaster (Dm). Analysis of immune signaling pathways and response modules reveals both conservative and rapidly evolving features associated with different functional gene categories and particular aspects of immune reactions. These dynamics reflect in part continuous readjustment between accommodation and rejection of pathogens and suggest how innate immunity may have evolved.


Asunto(s)
Aedes/genética , Anopheles/genética , Evolución Molecular , Inmunidad Innata/genética , Insectos Vectores/genética , Aedes/inmunología , Animales , Anopheles/inmunología , Péptidos Catiónicos Antimicrobianos/fisiología , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Drosophila melanogaster/genética , Drosophila melanogaster/inmunología , Genes de Insecto , Proteínas de Insectos/genética , Proteínas de Insectos/fisiología , Insectos Vectores/inmunología , Malaria/transmisión , Melaninas/metabolismo , Familia de Multigenes , Transducción de Señal , Especificidad de la Especie
20.
Immunol Rev ; 198: 127-48, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15199960

RESUMEN

In much of Africa, the mosquito Anopheles gambiae is the major vector of human malaria, a devastating infectious disease caused by Plasmodium parasites. Vector and parasite interact at multiple stages and locations, and the nature and effectiveness of this reciprocal interaction determines the success of transmission. Many of the interactions engage the mosquito's innate immunity, a primitive but very effective defense system. In some cases, the mosquito kills the parasite, thus blocking the transmission cycle. However, not all interactions are antagonistic; some represent immune evasion. The sequence of the A. gambiae genome revealed numerous potential components of the innate immune system, and it established that they evolve rapidly, as summarized in the present review. Their rapid evolution by gene family expansion diversification as well as the prevalence of haplotype alleles in the best-studied families may reflect selective adaptation of the immune system to the exigencies of multiple immune challenges in a variety of ecologic niches. As a follow-up to the comparative genomic analysis, the development of functional genomic methodologies has provided novel opportunities for understanding the immune system and the nature of its interactions with the parasite. In this context, identification of both Plasmodium antagonists and protectors in the mosquito represents a significant conceptual advance. In addition to providing fundamental understanding of primitive immune systems, studies of mosquito interactions with the parasite open unprecedented opportunities for novel interventions against malaria transmission. The generation of transgenic mosquitoes that resist malaria infection in the wild and the development of antimalarial 'smart sprays' capable of disrupting interactions that are protective of the parasite, or reinforcing others that are antagonistic, represent technical challenges but also immense opportunities for improvement of global health.


Asunto(s)
Anopheles/genética , Sistema Inmunológico , Inmunidad Innata , Insectos Vectores , Animales , Anopheles/inmunología , Anopheles/parasitología , Perfilación de la Expresión Génica , Genoma , Interacciones Huésped-Parásitos , Malaria/parasitología , Plasmodium berghei/genética , Plasmodium berghei/patogenicidad , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA