Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
PLoS Pathog ; 20(2): e1012008, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38354186

RESUMO

Leucine-rich repeat (LRR) proteins are commonly involved in innate immunity of animals and plants, including for pattern recognition of pathogen-derived elicitors. The Anopheles secreted LRR proteins APL1C and LRIM1 are required for malaria ookinete killing in conjunction with the complement-like TEP1 protein. However, the mechanism of parasite immune recognition by the mosquito remains unclear, although it is known that TEP1 lacks inherent binding specificity. Here, we find that APL1C and LRIM1 bind specifically to Plasmodium berghei ookinetes, even after depletion of TEP1 transcript and protein, consistent with a role for the LRR proteins in pathogen recognition. Moreover, APL1C does not bind to ookinetes of the human malaria parasite Plasmodium falciparum, and is not required for killing of this parasite, which correlates LRR binding specificity and immune protection. Most of the live P. berghei ookinetes that migrated into the extracellular space exposed to mosquito hemolymph, and almost all dead ookinetes, are bound by APL1C, thus associating LRR protein binding with parasite killing. We also find that APL1C binds to the surface of P. berghei sporozoites released from oocysts into the mosquito hemocoel and forms a potent barrier limiting salivary gland invasion and mosquito infectivity. Pathogen binding by APL1C provides the first functional explanation for the long-known requirement of APL1C for P. berghei ookinete killing in the mosquito midgut. We propose that secreted mosquito LRR proteins are required for pathogen discrimination and orientation of immune effector activity, potentially as functional counterparts of the immunoglobulin-based receptors used by vertebrates for antigen recognition.


Assuntos
Anopheles , Malária , Animais , Humanos , Proteínas de Repetições Ricas em Leucina , Anopheles/parasitologia , Esporozoítos/metabolismo , Proteínas/metabolismo , Plasmodium berghei/metabolismo
2.
BMC Genomics ; 20(1): 664, 2019 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-31429704

RESUMO

BACKGROUND: Mosquitoes are colonized by a large but mostly uncharacterized natural virome of RNA viruses, and the composition and distribution of the natural RNA virome may influence the biology and immunity of Anopheles malaria vector populations. RESULTS: Anopheles mosquitoes were sampled in malaria endemic forest village sites in Senegal and Cambodia, including Anopheles funestus, Anopheles gambiae group sp., and Anopheles coustani in Senegal, and Anopheles hyrcanus group sp., Anopheles maculatus group sp., and Anopheles dirus in Cambodia. The most frequent mosquito species sampled at both study sites are human malaria vectors. Small and long RNA sequences were depleted of mosquito host sequences, de novo assembled and clustered to yield non-redundant contigs longer than 500 nucleotides. Analysis of the assemblies by sequence similarity to known virus families yielded 115 novel virus sequences, and evidence supports a functional status for at least 86 of the novel viral contigs. Important monophyletic virus clades in the Bunyavirales and Mononegavirales orders were found in these Anopheles from Africa and Asia. The remaining non-host RNA assemblies that were unclassified by sequence similarity to known viruses were clustered by small RNA profiles, and 39 high-quality independent contigs strongly matched a pattern of classic RNAi processing of viral replication intermediates, suggesting they are entirely undescribed viruses. One thousand five hundred sixty-six additional high-quality unclassified contigs matched a pattern consistent with Piwi-interacting RNAs (piRNAs), suggesting that strand-biased piRNAs are generated from the natural virome in Anopheles. To functionally query piRNA effect, we analyzed piRNA expression in Anopheles coluzzii after infection with O'nyong nyong virus (family Togaviridae), and identified two piRNAs that appear to display specifically altered abundance upon arbovirus infection. CONCLUSIONS: Anopheles vectors of human malaria in Africa and Asia are ubiquitously colonized by RNA viruses, some of which are monophyletic but clearly diverged from other arthropod viruses. The interplay between small RNA pathways, immunity, and the virome may represent part of the homeostatic mechanism maintaining virome members in a commensal or nonpathogenic state, and could potentially influence vector competence.


Assuntos
Anopheles/virologia , Florestas , Mosquitos Vetores/virologia , Vírus de RNA/fisiologia , Animais , Anopheles/genética , Camboja , Regulação da Expressão Gênica , Mosquitos Vetores/genética , RNA Interferente Pequeno/genética , Senegal
3.
BMC Genomics ; 20(1): 698, 2019 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-31488060

RESUMO

Following the publication of this article [1], the authors reported that the original shading in columns 3 and 4 of Table 3, which indicated the presence or absence of viruses in each library, had been removed during typesetting.

4.
BMC Genomics ; 19(1): 526, 2018 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-29986645

RESUMO

BACKGROUND: Anopheles mosquitoes are efficient vectors of human malaria, but it is unknown why they do not transmit viruses as well as Aedes and Culex mosquitoes. The only arbovirus known to be consistently transmitted by Anopheles mosquitoes is O'nyong nyong virus (ONNV, genus Alphavirus, family Togaviridae). The interaction of Anopheles mosquitoes with RNA viruses has been relatively unexamined. RESULTS: We transcriptionally profiled the African malaria vector, Anopheles coluzzii, infected with ONNV. Mosquitoes were fed on an infectious bloodmeal and were analyzed by Illumina RNAseq at 3 days post-bloodmeal during the primary virus infection of the midgut epithelium, before systemic dissemination. Virus infection triggers transcriptional regulation of just 30 host candidate genes. Most of the regulated candidate genes are novel, without known function. Of the known genes, a significant cluster includes candidates with predicted involvement in carbohydrate metabolism. Two candidate genes encoding leucine-rich repeat immune (LRIM) factors point to possible involvement of immune protein complexes in the mosquito antiviral response. The primary ONNV infection by bloodmeal shares little transcriptional response in common with ONNV infection by intrathoracic injection, nor with midgut infection by the malaria parasites, Plasmodium falciparum or P. berghei. Profiling of A. coluzzii microRNA (miRNA) identified 118 known miRNAs and 182 potential novel miRNA candidates, with just one miRNA regulated by ONNV infection. This miRNA was not regulated by other previously reported treatments, and may be virus specific. Coexpression analysis of miRNA abundance and messenger RNA expression revealed discrete clusters of genes regulated by Imd and JAK/STAT, immune signaling pathways that are protective against ONNV in the primary infection. CONCLUSIONS: ONNV infection of the A. coluzzii midgut triggers a remarkably limited gene regulation program of mostly novel candidate genes, which likely includes host genes deployed for antiviral defense, as well as genes manipulated by the virus to facilitate infection. Functional dissection of the ONNV-response candidate genes is expected to generate novel insight into the mechanisms of virus-vector interaction.


Assuntos
Anopheles/genética , Arbovírus/patogenicidade , Mucosa Intestinal/metabolismo , Transcriptoma , Animais , Anopheles/metabolismo , Anopheles/virologia , Interações Hospedeiro-Patógeno/genética , Imunidade Inata/genética , Mucosa Intestinal/virologia , MicroRNAs/química , MicroRNAs/genética , MicroRNAs/metabolismo , Análise de Componente Principal , RNA/química , RNA/isolamento & purificação , RNA/metabolismo , Análise de Sequência de RNA
6.
Proc Natl Acad Sci U S A ; 112(2): E176-85, 2015 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-25548172

RESUMO

Arboviruses are transmitted by mosquitoes and other arthropods to humans and animals. The risk associated with these viruses is increasing worldwide, including new emergence in Europe and the Americas. Anopheline mosquitoes are vectors of human malaria but are believed to transmit one known arbovirus, o'nyong-nyong virus, whereas Aedes mosquitoes transmit many. Anopheles interactions with viruses have been little studied, and the initial antiviral response in the midgut has not been examined. Here, we determine the antiviral immune pathways of the Anopheles gambiae midgut, the initial site of viral infection after an infective blood meal. We compare them with the responses of the post-midgut systemic compartment, which is the site of the subsequent disseminated viral infection. Normal viral infection of the midgut requires bacterial flora and is inhibited by the activities of immune deficiency (Imd), JAK/STAT, and Leu-rich repeat immune factors. We show that the exogenous siRNA pathway, thought of as the canonical mosquito antiviral pathway, plays no detectable role in antiviral defense in the midgut but only protects later in the systemic compartment. These results alter the prevailing antiviral paradigm by describing distinct protective mechanisms in different body compartments and infection stages. Importantly, the presence of the midgut bacterial flora is required for full viral infectivity to Anopheles, in contrast to malaria infection, where the presence of the midgut bacterial flora is required for protection against infection. Thus, the enteric flora controls a reciprocal protection tradeoff in the vector for resistance to different human pathogens.


Assuntos
Anopheles/imunologia , Anopheles/virologia , Arbovírus/imunologia , Arbovírus/patogenicidade , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/transmissão , Animais , Anopheles/genética , Infecções por Arbovirus/imunologia , Infecções por Arbovirus/transmissão , Arbovírus/genética , Sistema Digestório/imunologia , Sistema Digestório/microbiologia , Sistema Digestório/virologia , Feminino , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Insetos Vetores/genética , Insetos Vetores/imunologia , Insetos Vetores/virologia , Janus Quinases/imunologia , Microbiota , Vírus O'nyong-nyong/genética , Vírus O'nyong-nyong/imunologia , Vírus O'nyong-nyong/patogenicidade , Plasmodium falciparum/imunologia , Plasmodium falciparum/patogenicidade , Interferência de RNA , RNA Interferente Pequeno/genética , Fatores de Transcrição STAT/imunologia , Transdução de Sinais/imunologia
7.
PLoS Pathog ; 11(12): e1005306, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26633695

RESUMO

Nucleotide variation patterns across species are shaped by the processes of natural selection, including exposure to environmental pathogens. We examined patterns of genetic variation in two sister species, Anopheles gambiae and Anopheles coluzzii, both efficient natural vectors of human malaria in West Africa. We used the differentiation signature displayed by a known coordinate selective sweep of immune genes APL1 and TEP1 in A. coluzzii to design a population genetic screen trained on the sweep, classified a panel of 26 potential immune genes for concordance with the signature, and functionally tested their immune phenotypes. The screen results were strongly predictive for genes with protective immune phenotypes: genes meeting the screen criteria were significantly more likely to display a functional phenotype against malaria infection than genes not meeting the criteria (p = 0.0005). Thus, an evolution-based screen can efficiently prioritize candidate genes for labor-intensive downstream functional testing, and safely allow the elimination of genes not meeting the screen criteria. The suite of immune genes with characteristics similar to the APL1-TEP1 selective sweep appears to be more widespread in the A. coluzzii genome than previously recognized. The immune gene differentiation may be a consequence of adaptation of A. coluzzii to new pathogens encountered in its niche expansion during the separation from A. gambiae, although the role, if any of natural selection by Plasmodium is unknown. Application of the screen allowed identification of new functional immune factors, and assignment of new functions to known factors. We describe biochemical binding interactions between immune proteins that underlie functional activity for malaria infection, which highlights the interplay between pathogen specificity and the structure of immune complexes. We also find that most malaria-protective immune factors display phenotypes for either human or rodent malaria, with broad specificity a rarity.


Assuntos
Anopheles/genética , Anopheles/imunologia , Insetos Vetores/genética , Insetos Vetores/imunologia , Animais , Sequência de Bases , Evolução Molecular , Genes de Insetos/imunologia , Variação Genética , Proteínas de Insetos/genética , Proteínas de Insetos/imunologia , Malária/transmissão , Camundongos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase
8.
Mol Ecol ; 25(7): 1494-510, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26846876

RESUMO

The recent discovery of a previously unknown genetic subgroup of Anopheles gambiae sensu lato underscores our incomplete understanding of complexities of vector population demographics in Anopheles. This subgroup, named GOUNDRY, does not rest indoors as adults and is highly susceptible to Plasmodium infection in the laboratory. Initial description of GOUNDRY suggested it differed from other known Anopheles taxa in surprising and sometimes contradictory ways, raising a number of questions about its age, population size and relationship to known subgroups. To address these questions, we sequenced the complete genomes of 12 wild-caught GOUNDRY specimens and compared these genomes to a panel of Anopheles genomes. We show that GOUNDRY is most closely related to Anopheles coluzzii, and the timing of cladogenesis is not recent, substantially predating the advent of agriculture. We find a large region of the X chromosome that has swept to fixation in GOUNDRY within the last 100 years, which may be an inversion that serves as a partial barrier to contemporary gene flow. Interestingly, we show that GOUNDRY has a history of inbreeding that is significantly associated with susceptibility to Plasmodium infection in the laboratory. Our results illuminate the genomic evolution of one of probably several cryptic, ecologically specialized subgroups of Anopheles and provide a potent example of how vector population dynamics may complicate efforts to control or eradicate malaria.


Assuntos
Anopheles/genética , Evolução Molecular , Genoma de Inseto , Plasmodium falciparum , Animais , Anopheles/parasitologia , Inversão Cromossômica , Fluxo Gênico , Especiação Genética , Genética Populacional , Endogamia , Insetos Vetores/genética , Insetos Vetores/parasitologia , Polimorfismo de Nucleotídeo Único , Dinâmica Populacional , Análise de Sequência de DNA , Cromossomo X/genética
9.
Malar J ; 15(1): 387, 2016 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-27456078

RESUMO

BACKGROUND: In many African countries malaria has declined sharply due to a synergy of actions marked by the introduction of vector control strategies, but the disease remains a leading cause of morbidity and mortality in Central African Republic (CAR). An entomological study was initiated with the aim to characterize the malaria vectors in Bangui, the capital of CAR, and determine their vector competence. METHODS: A cross-sectional entomological study was conducted in 15 sites of the district of Bangui, the capital of CAR, in September-October 2013 and a second collection was done in four of those sites between November and December 2013. Mosquitoes were collected by human landing catch (HLC) indoors and outdoors and by pyrethrum spray catch of indoor-resting mosquitoes. Mosquitoes were analysed for species and multiple other attributes, including the presence of Plasmodium falciparum circumsporozoite protein or DNA, blood meal source, 2La inversion karyotype, and the L1014F kdr insecticide resistance mutation. RESULTS: Overall, 1292 anophelines were analysed, revealing a predominance of Anopheles gambiae and Anopheles funestus, with a small fraction of Anopheles coluzzii. Molecular typing of the An. gambiae complex species showed that An. gambiae was predominant (95.7 %) as compared to An. coluzzii (2.1 %), and Anopheles arabiensis was not present. In some areas the involvement of secondary vectors, such as Anopheles coustani, expands the risk of infection. By HLC sampling, An. funestus displayed a stronger endophilic preference than mosquitoes from the An. gambiae sister taxa, with a mean indoor-capture rate of 54.3 % and 67.58 % for An. gambiae sister taxa and An. funestus, respectively. Human biting rates were measured overall for each of the species with 28 or 29 bites/person/night, respectively. Both vectors displayed a strong human feeding preference as determined by blood meal source, which was not different between the different sampling sites. An. coustani appears to be highly exophilic, with 92 % of HLC samples captured outdoors. The mean CSP rate in head-thorax sections of all Anopheles was 5.09 %, and was higher in An. gambiae s.l. (7.4 %) than in An. funestus (3.3 %). CSP-positive An. coustani were also detected in outdoor HLC samples. In the mosquitoes of the An. gambiae sister taxa the kdr-w mutant allele was nearly fixed, with 92.3 % resistant homozygotes, and no susceptible homozygotes detected. CONCLUSIONS: This study collected data on anopheline populations in CAR, behaviour of vectors and transmission levels. Further studies should investigate the biting behaviour and susceptibility status of the anophelines to different insecticides to allow the establishment of appropriate vector control based on practical entomological knowledge.


Assuntos
Anopheles/classificação , Anopheles/parasitologia , Biodiversidade , Mosquitos Vetores/classificação , Mosquitos Vetores/parasitologia , Plasmodium falciparum/isolamento & purificação , Animais , Anopheles/genética , República Centro-Africana , Estudos Transversais , Entomologia/métodos , Feminino , Resistência a Inseticidas , Mosquitos Vetores/genética
10.
Malar J ; 15: 191, 2016 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-27059057

RESUMO

BACKGROUND: Malaria remains a pervasive public health problem in sub-Saharan West Africa. Here mosquito vector populations were explored across four sites in Mali and the Republic of Guinea (Guinea Conakry). The study samples the major ecological zones of malaria-endemic regions in West Africa within a relatively small distance. METHODS: Mosquito vectors were sampled from larval pools, adult indoor resting sites, and indoor and outdoor human-host seeking adults. Mosquitoes were collected at sites spanning 350 km that represented arid savannah, humid savannah, semi-forest and deep forest ecological zones, in areas where little was previously known about malaria vector populations. 1425 mosquito samples were analysed by molecular assays to determine species, genetic attributes, blood meal sources and Plasmodium infection status. RESULTS: Anopheles gambiae and Anopheles coluzzii were the major anophelines represented in all collections across the ecological zones, with A. coluzzii predominant in the arid savannah and A. gambiae in the more humid sites. The use of multiple collection methodologies across the sampling sites allows assessment of potential collection bias of the different methods. The L1014F kdr insecticide resistance mutation (kdr-w) is found at high frequency across all study sites. This mutation appears to have swept almost to fixation, from low frequencies 6 years earlier, despite the absence of widespread insecticide use for vector control. Rates of human feeding are very high across ecological zones, with only small fractions of animal derived blood meals in the arid and humid savannah. About 30 % of freshly blood-fed mosquitoes were positive for Plasmodium falciparum presence, while the rate of mosquitoes with established infections was an order of magnitude lower. CONCLUSIONS: The study represents detailed vector characterization from an understudied area in West Africa with endemic malaria transmission. The deep forest study site includes the epicenter of the 2014 Ebola virus epidemic. With new malaria control interventions planned in Guinea, these data provide a baseline measure and an opportunity to assess the outcome of future interventions.


Assuntos
Anopheles/classificação , Anopheles/crescimento & desenvolvimento , Insetos Vetores , Plasmodium falciparum/isolamento & purificação , Animais , Anopheles/genética , Gâmbia , Guiné , Humanos , Mali
11.
BMC Genomics ; 16: 779, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26462916

RESUMO

BACKGROUND: The genome-wide association study (GWAS) techniques that have been used for genetic mapping in other organisms have not been successfully applied to mosquitoes, which have genetic characteristics of high nucleotide diversity, low linkage disequilibrium, and complex population stratification that render population-based GWAS essentially unfeasible at realistic sample size and marker density. METHODS: We designed a novel mapping strategy for the mosquito system that combines the power of linkage mapping with the resolution afforded by genetic association. We established founder colonies from West Africa, controlled for diversity, linkage disequilibrium and population stratification. Colonies were challenged by feeding on the infectious stage of the human malaria parasite, Plasmodium falciparum, mosquitoes were phenotyped for parasite load, and DNA pools for phenotypically similar mosquitoes were Illumina sequenced. Phenotype-genotype mapping was carried out in two stages, coarse and fine. RESULTS: In the first mapping stage, pooled sequences were analysed genome-wide for intervals displaying relativereduction in diversity between phenotype pools, and candidate genomic loci were identified for influence upon parasite infection levels. In the second mapping stage, focused genotyping of SNPs from the first mapping stage was carried out in unpooled individual mosquitoes and replicates. The second stage confirmed significant SNPs in a locus encoding two Toll-family proteins. RNAi-mediated gene silencing and infection challenge revealed that TOLL 11 protects mosquitoes against P. falciparum infection. CONCLUSIONS: We present an efficient and cost-effective method for genetic mapping using natural variation segregating in defined recent Anopheles founder colonies, and demonstrate its applicability for mapping in a complex non-model genome. This approach is a practical and preferred alternative to population-based GWAS for first-pass mapping of phenotypes in Anopheles. This design should facilitate mapping of other traits involved in physiology, epidemiology, and behaviour.


Assuntos
Anopheles/genética , Estudo de Associação Genômica Ampla , Malária Falciparum/genética , Plasmodium falciparum/genética , Receptores Toll-Like/genética , Animais , Anopheles/parasitologia , Mapeamento Cromossômico , Genoma de Inseto , Genótipo , Interações Hospedeiro-Parasita/genética , Humanos , Insetos Vetores/genética , Malária Falciparum/parasitologia , Malária Falciparum/transmissão , Fenótipo , Plasmodium falciparum/patogenicidade , Polimorfismo de Nucleotídeo Único
12.
Malar J ; 14: 391, 2015 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-26445487

RESUMO

BACKGROUND: Members of the Anopheles gambiae species complex are primary vectors of human malaria in Africa. It is known that a large haplotype shared between An. gambiae and Anopheles coluzzii by introgression carries point mutations of the voltage-gated sodium channel gene para, including the L1014F kdr mutation associated with insensitivity to pyrethroid insecticides. Carriage of L1014F kdr is also correlated with higher susceptibility to infection with Plasmodium falciparum. However, the genetic mechanism and causative gene(s) underlying the parasite susceptibility phenotype are not known. METHODS: Mosquitoes from the wild Burkina Faso population were challenged by feeding on natural P. falciparum gametocytes. Oocyst infection phenotypes were determined and were tested for association with SNP genotypes. Candidate genes in the detected locus were prioritized and RNAi-mediated gene silencing was used to functionally test for gene effects on P. falciparum susceptibility. RESULTS: A genetic locus, Pfin6, was identified that influences infection levels of P. falciparum in mosquitoes. The locus segregates as a ~3 Mb haplotype carrying 65 predicted genes including the para gene. The haplotype carrying the kdr allele of para is linked to increased parasite infection prevalence, but many single nucleotide polymorphisms on the haplotype are also equally linked to the infection phenotype. Candidate genes in the haplotype were prioritized and functionally tested. Silencing of para did not influence P. falciparum infection, while silencing of a predicted immune gene, serine protease ClipC9, allowed development of significantly increased parasite numbers. CONCLUSIONS: Genetic variation influencing Plasmodium infection in wild Anopheles is linked to a natural ~3 megabase haplotype on chromosome 2L that carries the kdr allele of the para gene. Evidence suggests that para gene function does not directly influence parasite susceptibility, and the association of kdr with infection may be due to tight linkage of kdr with other gene(s) on the haplotype. Further work will be required to determine if ClipC9 influences the outcome of P. falciparum infection in nature, as well as to confirm the absence of a direct influence by para.


Assuntos
Anopheles/genética , Anopheles/parasitologia , Loci Gênicos , Haplótipos , Resistência a Inseticidas , Plasmodium falciparum/crescimento & desenvolvimento , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Animais , Anopheles/imunologia , Burkina Faso , Feminino , Ligação Genética , Plasmodium falciparum/imunologia
13.
Blood ; 119(24): e172-80, 2012 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-22517905

RESUMO

Achievement of malaria elimination requires development of novel strategies interfering with parasite transmission, including targeting the parasite sexual stages (gametocytes). The formation of Plasmodium falciparum gametocytes in the human host takes several days during which immature gametocyte-infected erythrocytes (GIEs) sequester in host tissues. Only mature stage GIEs circulate in the peripheral blood, available to uptake by the Anopheles vector. Mechanisms underlying GIE sequestration and release in circulation are virtually unknown. We show here that mature GIEs are more deformable than immature stages using ektacytometry and microsphiltration methods, and that a switch in cellular deformability in the transition from immature to mature gametocytes is accompanied by the deassociation of parasite-derived STEVOR proteins from the infected erythrocyte membrane. We hypothesize that mechanical retention contributes to sequestration of immature GIEs and that regained deformability of mature gametocytes is associated with their release in the bloodstream and ability to circulate. These processes are proposed to play a key role in P falciparum gametocyte development in the host and to represent novel and unconventional targets for interfering with parasite transmission.


Assuntos
Deformação Eritrocítica/fisiologia , Eritrócitos/parasitologia , Estágios do Ciclo de Vida , Malária Falciparum/sangue , Malária Falciparum/transmissão , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/fisiologia , Adulto , Animais , Antígenos de Protozoários/metabolismo , Imunofluorescência , Humanos , Malária Falciparum/parasitologia , Plasmodium falciparum/ultraestrutura , Transporte Proteico
14.
PLoS Biol ; 9(3): e1000600, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21408087

RESUMO

The three-gene APL1 locus encodes essential components of the mosquito immune defense against malaria parasites. APL1 was originally identified because it lies within a mapped QTL conferring the vector mosquito Anopheles gambiae natural resistance to the human malaria parasite, Plasmodium falciparum, and APL1 genes have subsequently been shown to be involved in defense against several species of Plasmodium. Here, we examine molecular population genetic variation at the APL1 gene cluster in spatially and temporally diverse West African collections of A. gambiae. The locus is extremely polymorphic, showing evidence of adaptive evolutionary maintenance of genetic variation. We hypothesize that this variability aids in defense against genetically diverse pathogens, including Plasmodium. Variation at APL1 is highly structured across geographic and temporal subpopulations. In particular, diversity is exceptionally high during the rainy season, when malaria transmission rates are at their peak. Much less allelic diversity is observed during the dry season when mosquito population sizes and malaria transmission rates are low. APL1 diversity is weakly stratified by the polymorphic 2La chromosomal inversion but is very strongly subdivided between the M and S "molecular forms." We find evidence that a recent selective sweep has occurred at the APL1 locus in M form mosquitoes only. The independently reported observation of a similar M-form restricted sweep at the Tep1 locus, whose product physically interacts with APL1C, suggests that epistatic selection may act on these two loci causing them to sweep coordinately.


Assuntos
Anopheles/genética , Proteínas de Insetos/genética , Insetos Vetores/genética , Plasmodium falciparum/imunologia , Polimorfismo Genético , Seleção Genética , Adaptação Biológica , Animais , Anopheles/imunologia , Anopheles/parasitologia , Evolução Molecular , Geografia , Imunidade Inata/genética , Proteínas de Insetos/química , Insetos Vetores/parasitologia , Estações do Ano
15.
Malar J ; 12: 67, 2013 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-23421809

RESUMO

BACKGROUND: Malaria can be caused by five Plasmodium species. Due to their higher prevalence, much of the research concentrates on Plasmodium falciparum and Plasmodium vivax. In Burkina Faso, where P. falciparum co-exists with Plasmodium malariae and Plasmodium ovale, there is not much data about the prevalence of the latter two species across human population. Moreover, interactions between co-infecting Plasmodium species are not documented. The aim of the current research is to determine species-specific prevalence and temporal distribution. The potential interactions between co-infecting Plasmodium species amongst the child-aged population in Burkina Faso are also discussed. METHODS: The study took place in the Sudanese savannah zone in Burkina Faso in a rural village, Laye. Surveys were conducted during the wet season across four years, 2007 to 2010. Volunteers aged three to 15 years with parental signed consent were enrolled. Ten children per week were screened for any history of pain, fever. Parasitological data were obtained by blood slide processing. RESULTS: Three sympatric Plasmodium species were recorded during this study with an average prevalence of 70.7%. Species temporal distribution showed an increase of P. malariae parasite prevalence from 0.9% in 2007 to 13.2% in 2010. Within a season, P. falciparum occurred in the overall study period while P. malariae and P. ovale were highly prevalent after the rainy part of this period. Species-specific infection analysis showed that in a comparison of mono-infections, P. malariae gametocyte prevalence and median density were higher than those of P. falciparum (88.9% vs 34.5% and 124.0 vs 40.0 gametocytes/µl, respectively). Likewise, in P. falciparum co-infections with P. malariae or P. ovale, gametocyte prevalence was also higher than in P. falciparum mono-infection. However, in P. falciparum mixed infection with P. malariae, P. falciparum gametocyte prevalence and median density as well as asexual form density decreased compared to P. falciparum mono-infection while for P. malariae mono-infection, only asexual form density significantly vary. CONCLUSION: These data revealed high gametocyte prevalence in other Plasmodium species than P. falciparum with a significant variation of P. malariae gametocyte carriers and gametocyte density across years. Molecular tools and entomological studies are needed to highly assess species-specific contribution to malaria transmission.


Assuntos
Malária/epidemiologia , Malária/parasitologia , Plasmodium falciparum/isolamento & purificação , Plasmodium malariae/isolamento & purificação , Plasmodium ovale/isolamento & purificação , Animais , Burkina Faso/epidemiologia , Criança , Coinfecção/epidemiologia , Coinfecção/parasitologia , Feminino , Humanos , Masculino , Carga Parasitária , Prevalência , População Rural
16.
Malar J ; 12: 204, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23764031

RESUMO

BACKGROUND: The Anopheles gambiae sensu lato (s.l.) species complex in Burkina Faso consists of Anopheles arabiensis, and molecular forms M and S of Anopheles gambiae sensu stricto (s.s.). Previous studies comparing the M and S forms for level of infection with Plasmodium falciparum have yielded conflicting results. METHODS: Mosquito larvae were sampled from natural pools, reared to adulthood under controlled conditions, and challenged with natural P. falciparum by experimental feeding with blood from gametocyte carriers. Oocyst infection prevalence and intensity was determined one week after infection. DNA from carcasses was genotyped to identify species and molecular form. RESULTS: In total, 7,400 adult mosquitoes grown from wild-caught larvae were challenged with gametocytes in 29 experimental infections spanning four transmission seasons. The overall infection prevalence averaged 40.7% for A. gambiae M form, 41.4% for A. gambiae S form, and 40.1% for A. arabiensis. There was no significant difference in infection prevalence or intensity between the three population groups. Notably, infection experiments in which the population groups were challenged in parallel on the same infective blood displayed less infection difference between population groups, while infections with less balanced composition of population groups had lower statistical power and displayed apparent differences that fluctuated more often from the null average. CONCLUSION: The study clearly establishes that, at the study site in Burkina Faso, there is no difference in genetic susceptibility to P. falciparum infection between three sympatric population groups of the A. gambiae s.l. complex. Feeding the mosquito groups on the same infective blood meal greatly increases statistical power. Conversely, comparison of the different mosquito groups between, rather than within, infections yields larger apparent difference between mosquito groups, resulting from lower statistical power and greater noise, and could lead to false-positive results. In making infection comparisons between population groups, it is more accurate to compare the different groups after feeding simultaneously upon the same infective blood.


Assuntos
Anopheles/fisiologia , Anopheles/parasitologia , Plasmodium falciparum/crescimento & desenvolvimento , Animais , Burkina Faso , DNA de Protozoário/genética , DNA de Protozoário/isolamento & purificação , Genótipo , Plasmodium falciparum/classificação , Plasmodium falciparum/genética , Plasmodium falciparum/isolamento & purificação
17.
Viruses ; 15(3)2023 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-36992298

RESUMO

Mosquito-borne diseases caused by viruses and parasites are responsible for more than 700 million infections each year. Anopheles and Aedes are the two major vectors for, respectively, malaria and arboviruses. Anopheles mosquitoes are the primary vector of just one known arbovirus, the alphavirus o'nyong-nyong virus (ONNV), which is closely related to the chikungunya virus (CHIKV), vectored by Aedes mosquitoes. However, Anopheles harbor a complex natural virome of RNA viruses, and a number of pathogenic arboviruses have been isolated from Anopheles mosquitoes in nature. CHIKV and ONNV are in the same antigenic group, the Semliki Forest virus complex, are difficult to distinguish via immunodiagnostic assay, and symptomatically cause essentially the same human disease. The major difference between the arboviruses appears to be their differential use of mosquito vectors. The mechanisms governing this vector specificity are poorly understood. Here, we summarize intrinsic and extrinsic factors that could be associated with vector specificity by these viruses. We highlight the complexity and multifactorial aspect of vectorial specificity of the two alphaviruses, and evaluate the level of risk of vector shift by ONNV or CHIKV.


Assuntos
Aedes , Anopheles , Arbovírus , Febre de Chikungunya , Vírus Chikungunya , Animais , Humanos , Vírus O'nyong-nyong/genética , Mosquitos Vetores , Vírus Chikungunya/genética
18.
Sci Rep ; 13(1): 14572, 2023 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-37666840

RESUMO

The voltage-gated sodium channel, para, is a target of DDT and pyrethroid class insecticides. Single nucleotide mutations in para, called knockdown resistant or kdr, which contribute to resistance against DDT and pyrethroid insecticides, have been correlated with increased susceptibility of Anopheles to the human malaria parasite Plasmodium falciparum. However, a direct role of para activity on Plasmodium infection has not yet been established. Here, using RNA-mediated silencing, we provide in vivo direct evidence for the requirement of wild-type (wt) para function for insecticide activity of deltamethrin. Depletion of wt para, which is susceptible to insecticide, causes deltamethrin tolerance, indicating that insecticide-resistant kdr alleles are likely phenocopies of loss of para function. We then show that normal para activity in An. coluzzii limits Plasmodium infection prevalence for both P. falciparum and P. berghei. A transcriptomic analysis revealed that para activity does not modulate the expression of immune genes. However, loss of para function led to enteric dysbiosis with a significant increase in the total bacterial abundance, and we show that para function limiting Plasmodium infection is microbiota dependent. In the context of the bidirectional "enteric microbiota-brain" axis studied in mammals, these results pave the way for studying whether the activity of the nervous system could control Anopheles vector competence.


Assuntos
Anopheles , Inseticidas , Malária Falciparum , Microbiota , Canais de Sódio Disparados por Voltagem , Humanos , Animais , Anopheles/genética , DDT , Mosquitos Vetores/genética , Canais de Sódio Disparados por Voltagem/genética , Mamíferos
19.
J Biol Chem ; 286(47): 40824-34, 2011 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-21965675

RESUMO

Mosquitoes transmit Plasmodium and certain arboviruses during blood feeding, when they are injected along with saliva. Mosquito saliva interferes with the host's hemostasis and inflammation response and influences the transmission success of some pathogens. One family of mosquito salivary gland proteins, named SGS, is composed of large bacterial-type proteins that in Aedes aegypti were implicated as receptors for Plasmodium on the basal salivary gland surface. Here, we characterize the biology of two SGSs in the malaria mosquito, Anopheles gambiae, and demonstrate their involvement in blood feeding. Western blots and RT-PCR showed that Sgs4 and Sgs5 are produced exclusively in female salivary glands, that expression increases with age and after blood feeding, and that protein levels fluctuate in a circadian manner. Immunohistochemistry showed that SGSs are present in the acinar cells of the distal lateral lobes and in the salivary ducts of the proximal lobes. SDS-PAGE, Western blots, bite blots, and immunization via mosquito bites showed that SGSs are highly immunogenic and form major components of mosquito saliva. Last, Western and bioinformatic analyses suggest that SGSs are secreted via a non-classical pathway that involves cleavage into a 300-kDa soluble fragment and a smaller membrane-bound fragment. Combined, these data strongly suggest that SGSs play an important role in blood feeding. Together with their role in malaria transmission, we propose that SGSs could be used as markers of human exposure to mosquito bites and in the development of disease control strategies.


Assuntos
Anopheles/imunologia , Anopheles/metabolismo , Proteínas do Grude Salivar de Drosophila/metabolismo , Fatores Imunológicos/metabolismo , Saliva/imunologia , Saliva/metabolismo , Glândulas Salivares/metabolismo , Animais , Biologia Computacional , Proteínas de Drosophila , Feminino , Regulação da Expressão Gênica , Proteínas do Grude Salivar de Drosophila/genética , Proteínas do Grude Salivar de Drosophila/imunologia , Humanos , Fatores Imunológicos/genética , Fatores Imunológicos/imunologia , Proteólise , Fatores de Tempo
20.
Sci Rep ; 12(1): 6315, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35428783

RESUMO

Entomopathogenic fungi have been explored as a potential biopesticide to counteract the insecticide resistance issue in mosquitoes. However, little is known about the possibility that genetic resistance to fungal biopesticides could evolve in mosquito populations. Here, we detected an important genetic component underlying Anopheles coluzzii survival after exposure to the entomopathogenic fungus Metarhizium anisopliae. A familiality study detected variation for survival among wild mosquito isofemale pedigrees, and genetic mapping identified two loci that significantly influence mosquito survival after fungus exposure. One locus overlaps with a previously reported locus for Anopheles susceptibility to the human malaria parasite Plasmodium falciparum. Candidate gene studies revealed that two LRR proteins encoded by APL1C and LRIM1 genes in this newly mapped locus are required for protection of female A. coluzzii from M. anisopliae, as is the complement-like factor Tep1. These results indicate that natural Anopheles populations already segregate frequent genetic variation for differential mosquito survival after fungal challenge and suggest a similarity in Anopheles protective responses against fungus and Plasmodium. However, this immune similarity raises the possibility that fungus-resistant mosquitoes could also display enhanced resistance to Plasmodium, suggesting an advantage of selecting for fungus resistance in vector populations to promote naturally diminished malaria vector competence.


Assuntos
Anopheles , Malária , Metarhizium , Plasmodium , Animais , Anopheles/parasitologia , Feminino , Humanos , Metarhizium/genética , Mosquitos Vetores/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA