Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 136(5): 805-9, 2009 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-19269357

RESUMO

In 2007, the European Research Council (ERC) was launched amid much fanfare with the goal of spearheading Europe's aspirations to become the most dynamic and competitive knowledge-based society in the world. Here, we examine the results of the first two ERC calls for research grants and discuss the latest developments and the challenges that face this unique research council.


Assuntos
Pesquisa , Europa (Continente) , Pesquisa/economia , Apoio à Pesquisa como Assunto , Sociedades Científicas/economia , Sociedades Científicas/organização & administração
2.
PLoS Pathog ; 9(1): e1003145, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23382679

RESUMO

Reverse genetics in the mosquito Anopheles gambiae by RNAi mediated gene silencing has led in recent years to an advanced understanding of the mosquito immune response against infections with bacteria and malaria parasites. We developed RNAi screens in An. gambiae hemocyte-like cells using a library of double-stranded RNAs targeting 109 genes expressed highly or specifically in mosquito hemocytes to identify novel regulators of the hemocyte immune response. Assays included phagocytosis of bacterial bioparticles, expression of the antimicrobial peptide CEC1, and basal and induced expression of the mosquito complement factor LRIM1. A cell viability screen was also carried out to assess dsRNA cytotoxicity and to identify genes involved in cell growth and survival. Our results identify 22 novel immune regulators, including proteins putatively involved in phagosome assembly and maturation (Ca²âº channel, v-ATPase and cyclin-dependent protein kinase), pattern recognition (fibrinogen-domain lectins and Nimrod), immune modulation (peptidase and serine protease homolog), immune signaling (Eiger and LPS-induced factor), cell adhesion and communication (Laminin B1 and Ninjurin) and immune homeostasis (Lipophorin receptor). The development of robust functional cell-based assays paves the way for genome-wide functional screens to study the mosquito immune response to infections with human pathogens.


Assuntos
Anopheles/genética , Proteínas do Sistema Complemento/genética , Inativação Gênica , Hemócitos/imunologia , Imunidade Ativa/genética , Animais , Anopheles/imunologia , Sobrevivência Celular , Células Cultivadas , Proteínas do Sistema Complemento/imunologia , Escherichia coli , Expressão Gênica , Estudo de Associação Genômica Ampla , Hemócitos/citologia , Hemócitos/microbiologia , Interações Hospedeiro-Patógeno , Proteínas de Insetos , Oocistos/citologia , Oocistos/imunologia , Fagocitose/fisiologia , Interferência de RNA , RNA de Cadeia Dupla/farmacologia , RNA Interferente Pequeno
3.
Proc Natl Acad Sci U S A ; 106(50): 21270-5, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19940242

RESUMO

Insect hemocytes mediate important cellular immune responses including phagocytosis and encapsulation and also secrete immune factors such as opsonins, melanization factors, and antimicrobial peptides. However, the molecular composition of these important immune cells has not been elucidated in depth, because of their scarcity in the circulating hemolymph, their adhesion to multiple tissues and the lack of primary culture methods to produce sufficient material for a genome-wide analysis. In this study, we report a genome-wide molecular characterization of circulating hemocytes collected from the hemolymph of adult female Anopheles gambiae mosquitoes--the major mosquito vector of human malaria in subSaharan Africa. Their molecular profile identified 1,485 transcripts with enriched expression in these cells, and many of these genes belong to innate immune gene families. This hemocyte-specific transcriptome is compared to those of Drosophila melanogaster and two other mosquitoes, Aedes aegypti and Armigeres subalbatus. We report the identification of two genes as ubiquitous hemocyte markers and several others as hemocyte subpopulation markers. We assess, via an RNAi screen, the roles in development of Plasmodium berghei of 63 genes expressed in hemocytes and provide a molecular comparison of the transcriptome of these cells during malaria infection.


Assuntos
Anopheles/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Genoma de Inseto/genética , Hemócitos/metabolismo , Aedes/genética , Animais , Drosophila melanogaster/genética , Feminino , Marcadores Genéticos , Malária , Plasmodium , RNA Mensageiro/análise
4.
Infect Immun ; 79(11): 4708-15, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21844236

RESUMO

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.


Assuntos
Anopheles/imunologia , Anopheles/parasitologia , Insetos Vetores/imunologia , Insetos Vetores/parasitologia , Plasmodium falciparum/fisiologia , Animais , Anopheles/genética , Anopheles/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Interações Hospedeiro-Parasita , Humanos , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Insetos Vetores/genética , Insetos Vetores/metabolismo , Camundongos , Filogenia , Plasmodium berghei/fisiologia
5.
PLoS Pathog ; 5(8): e1000542, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19662170

RESUMO

Recognition of peptidoglycan (PGN) is paramount for insect antibacterial defenses. In the fruit fly Drosophila melanogaster, the transmembrane PGN Recognition Protein LC (PGRP-LC) is a receptor of the Imd signaling pathway that is activated after infection with bacteria, mainly Gram-negative (Gram-). Here we demonstrate that bacterial infections of the malaria mosquito Anopheles gambiae are sensed by the orthologous PGRPLC protein which then activates a signaling pathway that involves the Rel/NF-kappaB transcription factor REL2. PGRPLC signaling leads to transcriptional induction of antimicrobial peptides at early stages of hemolymph infections with the Gram-positive (Gram+) bacterium Staphylococcus aureus, but a different signaling pathway might be used in infections with the Gram- bacterium Escherichia coli. The size of mosquito symbiotic bacteria populations and their dramatic proliferation after a bloodmeal, as well as intestinal bacterial infections, are also controlled by PGRPLC signaling. We show that this defense response modulates mosquito infection intensities with malaria parasites, both the rodent model parasite, Plasmodium berghei, and field isolates of the human parasite, Plasmodium falciparum. We propose that the tripartite interaction between mosquito microbial communities, PGRPLC-mediated antibacterial defense and infections with Plasmodium can be exploited in future interventions aiming to control malaria transmission. Molecular analysis and structural modeling provided mechanistic insights for the function of PGRPLC. Alternative splicing of PGRPLC transcripts produces three main isoforms, of which PGRPLC3 appears to have a key role in the resistance to bacteria and modulation of Plasmodium infections. Structural modeling indicates that PGRPLC3 is capable of binding monomeric PGN muropeptides but unable to initiate dimerization with other isoforms. A dual role of this isoform is hypothesized: it sequesters monomeric PGN dampening weak signals and locks other PGRPLC isoforms in binary immunostimulatory complexes further enhancing strong signals.


Assuntos
Anopheles/imunologia , Anopheles/microbiologia , Infecções Bacterianas/imunologia , Proteínas de Transporte/imunologia , Plasmodium/imunologia , Processamento Alternativo , Sequência de Aminoácidos , Animais , Anopheles/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/genética , DNA Bacteriano/genética , Feminino , Malária/imunologia , Malária/transmissão , Dados de Sequência Molecular , Isoformas de Proteínas/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia
6.
PLoS Pathog ; 5(8): e1000539, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19662167

RESUMO

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.


Assuntos
Culicidae/parasitologia , Insetos Vetores/parasitologia , Plasmodium berghei/crescimento & desenvolvimento , Proteínas de Protozoários/genética , Sequência de Aminoácidos , Animais , Southern Blotting , Feminino , Imunofluorescência , Expressão Gênica , Regulação da Expressão Gênica , Genes de Protozoários/genética , Malária/transmissão , Masculino , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Filogenia , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Proteínas de Protozoários/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Nucleic Acids Res ; 37(Database issue): D583-7, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19028744

RESUMO

VectorBase (http://www.vectorbase.org) is an NIAID-funded Bioinformatic Resource Center focused on invertebrate vectors of human pathogens. VectorBase annotates and curates vector genomes providing a web accessible integrated resource for the research community. Currently, VectorBase contains genome information for three mosquito species: Aedes aegypti, Anopheles gambiae and Culex quinquefasciatus, a body louse Pediculus humanus and a tick species Ixodes scapularis. Since our last report VectorBase has initiated a community annotation system, a microarray and gene expression repository and controlled vocabularies for anatomy and insecticide resistance. We have continued to develop both the software infrastructure and tools for interrogating the stored data.


Assuntos
Vetores Artrópodes/genética , Culicidae/genética , Bases de Dados Genéticas , Aedes/genética , Animais , Anopheles/genética , Culex/genética , Culicidae/metabolismo , Perfilação da Expressão Gênica , Genoma de Inseto , Genômica , Ixodes/genética , Pediculus/genética , Vocabulário Controlado
8.
PLoS Pathog ; 4(5): e1000069, 2008 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-18483558

RESUMO

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.


Assuntos
Anopheles/parasitologia , Vetores de Doenças , Malária Falciparum/parasitologia , Plasmodium berghei/fisiologia , Plasmodium falciparum/fisiologia , Animais , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Apolipoproteína A-II/genética , Apolipoproteína A-II/metabolismo , Criança , Pré-Escolar , DNA de Protozoário/análise , Sistema Digestório/parasitologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Inativação Gênica , Predisposição Genética para Doença , Interações Hospedeiro-Parasita , Humanos , Malária Falciparum/sangue , Malária Falciparum/transmissão , Oocistos/fisiologia , Plasmodium berghei/patogenicidade , Plasmodium falciparum/patogenicidade , Polimorfismo de Nucleotídeo Único , Transcrição Gênica , Proteína da Síndrome de Wiskott-Aldrich/genética , Proteína da Síndrome de Wiskott-Aldrich/metabolismo
9.
Curr Opin Genet Dev ; 16(4): 384-91, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16793259

RESUMO

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.


Assuntos
Culicidae/parasitologia , Plasmodium/crescimento & desenvolvimento , Animais , Movimento , Oocistos/crescimento & desenvolvimento , Plasmodium/fisiologia , Glândulas Salivares/parasitologia , Esporozoítos/crescimento & desenvolvimento
10.
Cell Microbiol ; 10(4): 891-8, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18005239

RESUMO

For malaria transmission to occur, Plasmodium sporozoites must infect the salivary glands of their mosquito vectors. This study reports that Anopheles gambiae SRPN6 participates in a local salivary gland epithelial response against the rodent malaria parasite, Plasmodium berghei. We showed previously that SRPN6, an immune inducible midgut invasion marker, influences ookinete development. Here we report that SRPN6 is also specifically induced in salivary glands with the onset of sporozoite invasion. The protein is located in the basal region of epithelial cells in proximity to invading sporozoites. Knockdown of SRPN6 during the late phase of sporogony by RNAi has no effect on oocyst rupture but significantly increases the number of sporozoites present in salivary glands. Despite several differences between the passage of Plasmodium through the midgut and the salivary glands, this study identifies a striking overlap in the molecular responses of these two epithelia to parasite invasion.


Assuntos
Anopheles/parasitologia , Proteínas de Insetos/fisiologia , Plasmodium berghei/crescimento & desenvolvimento , Glândulas Salivares/parasitologia , Esporozoítos/crescimento & desenvolvimento , Animais , Anopheles/genética , Anopheles/metabolismo , Western Blotting , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Insetos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Glândulas Salivares/metabolismo
11.
Nucleic Acids Res ; 35(Database issue): D503-5, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17145709

RESUMO

VectorBase (http://www.vectorbase.org/) is a web-accessible data repository for information about invertebrate vectors of human pathogens. VectorBase annotates and maintains vector genomes providing an integrated resource for the research community. Currently, VectorBase contains genome information for two organisms: Anopheles gambiae, a vector for the Plasmodium protozoan agent causing malaria, and Aedes aegypti, a vector for the flaviviral agents causing Yellow fever and Dengue fever.


Assuntos
Aedes/genética , Anopheles/genética , Bases de Dados Genéticas , Genoma de Inseto , Insetos Vetores/genética , Animais , Sequência de Bases , Sequência Conservada , Genômica , Humanos , Internet , Interface Usuário-Computador
12.
BMC Genomics ; 9: 227, 2008 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-18489733

RESUMO

BACKGROUND: Anopheles innate immunity affects Plasmodium development and is a potential target of innovative malaria control strategies. The extent and distribution of nucleotide diversity in immunity genes might provide insights into the evolutionary forces that condition pathogen-vector interactions. The discovery of polymorphisms is an essential step towards association studies of susceptibility to infection. RESULTS: We sequenced coding fragments of 72 immune related genes in natural populations of Anopheles gambiae and of 37 randomly chosen genes to provide a background measure of genetic diversity across the genome. Mean nucleotide diversity (pi) was 0.0092 in the A. gambiae S form, 0.0076 in the M form and 0.0064 in A. arabiensis. Within each species, no statistically significant differences in mean nucleotide diversity were detected between immune related and non immune related genes. Strong purifying selection was detected in genes of both categories, presumably reflecting strong functional constraints. CONCLUSION: Our results suggest similar patterns and rates of molecular evolution in immune and non-immune genes in A. gambiae. The 3,214 Single Nucleotide Polymorphisms (SNPs) that we identified are the first large set of Anopheles SNPs from fresh, field-collected material and are relevant markers for future phenotype-association studies.


Assuntos
Anopheles/genética , Anopheles/imunologia , Evolução Molecular , Imunidade Inata/genética , Polimorfismo de Nucleotídeo Único , Animais , Genes de Insetos , Variação Genética , Humanos , Insetos Vetores/genética , Insetos Vetores/imunologia , Insetos Vetores/parasitologia , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Plasmodium falciparum/imunologia , Plasmodium falciparum/patogenicidade , Seleção Genética , Especificidade da Espécie
13.
Curr Biol ; 15(13): 1185-95, 2005 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-16005290

RESUMO

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.


Assuntos
Anopheles/genética , Anopheles/parasitologia , Sistema Digestório/parasitologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Plasmodium berghei , Actinas/metabolismo , Animais , Anopheles/metabolismo , Apoptose/genética , Análise por Conglomerados , Epitélio/metabolismo , Epitélio/parasitologia , Genômica/métodos , Imunidade Inata/genética , Análise de Sequência com Séries de Oligonucleotídeos , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Nat Rev Microbiol ; 3(3): 262-8, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15703759

RESUMO

Diseases that are transmitted by arthropods cause severe morbidity and mortality throughout the world. The burden of many of these diseases is borne largely by developing countries. Advances in vector genomics offer new promise for the control of arthropod vectors of disease. Radical changes in vector-biology research are required if scientists are to exploit genomic data and implement changes in public health.


Assuntos
Vetores Artrópodes/genética , Controle de Doenças Transmissíveis/métodos , Genômica , Animais , Vetores Artrópodes/microbiologia , Vetores Artrópodes/parasitologia , Doenças Transmissíveis/mortalidade , Doenças Transmissíveis/transmissão , Humanos
15.
Curr Opin Microbiol ; 8(4): 415-21, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15996894

RESUMO

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.


Assuntos
Anopheles/parasitologia , Regulação da Expressão Gênica no Desenvolvimento , Insetos Vetores/parasitologia , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/fisiologia , Animais , Interações Hospedeiro-Parasita , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo
16.
Insect Biochem Mol Biol ; 36(4): 282-90, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16551542

RESUMO

Mosquitoes use effective immune responses, including phagocytosis, to fight microbial infection. Here we show that in an Anopheles gambiae immune responsive cell line, RGD recognizing receptors play an important role in the phagocytic response, suggesting overlap between molecular components implicated in adhesion and phagocytosis. Integrins are a major class of adhesive receptors that recognize ligands containing an RGD motif. We have cloned a gene encoding a new beta integrin, BINT2, and demonstrated its involvement in Escherichia coli engulfment. Based on molecular modeling, we propose a structural reason for the role of BINT2, but not BINT1, on phagocytosis of Gram-negative bacteria. Using bioinformatic tools, we have identified and compared the complete A. gambiae integrin repertoire as a prelude to a future systematic functional study.


Assuntos
Anopheles/metabolismo , Anopheles/microbiologia , Escherichia coli/fisiologia , Proteínas de Insetos/fisiologia , Cadeias beta de Integrinas/fisiologia , Fagocitose/fisiologia , Animais , Anopheles/citologia , Adesão Celular/fisiologia , Linhagem Celular , Clonagem Molecular , Cristalografia por Raios X , Proteínas de Insetos/química , Proteínas de Insetos/metabolismo , Cadeias beta de Integrinas/química , Cadeias beta de Integrinas/metabolismo , Modelos Moleculares , Filogenia , Receptores Imunológicos/fisiologia , Receptores de Peptídeos/fisiologia , Staphylococcus aureus/fisiologia
17.
Mol Biochem Parasitol ; 142(1): 76-87, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15907562

RESUMO

Simultaneous microarray-based transcription analysis of 4987 Anopheles stephensi midgut and Plasmodium berghei infection stage specific cDNAs was done at seven successive time points: 6, 20 and 40h, and 4, 8, 14 and 20 days after ingestion of malaria infected blood. The study reveals the molecular components of several Anopheles processes relating to blood digestion, midgut expansion and response to Plasmodium-infected blood such as digestive enzymes, transporters, cytoskeletal and structural components and stress and immune responsive factors. In parallel, the analysis provide detailed expression patterns of Plasmodium genes encoding essential developmental and metabolic factors and proteins implicated in interaction with the mosquito vector and vertebrate host such as kinases, transcription and translational factors, cytoskeletal components and a variety of surface proteins, some of which are potent vaccine targets. Temporal correlation between transcription profiles of both organisms identifies putative gene clusters of interacting processes, such as Plasmodium invasion of the midgut epithelium, Anopheles immune responses to Plasmodium infection, and apoptosis and expulsion of invaded midgut cells from the epithelium. Intriguing transcription patterns for highly variable Plasmodium surface antigens may indicate parasite strategies to avoid recognition by the mosquito's immune surveillance system.


Assuntos
Anopheles/parasitologia , Proteínas de Insetos/metabolismo , Plasmodium berghei/patogenicidade , Proteoma , Proteínas de Protozoários/metabolismo , Transcrição Gênica , Animais , Anopheles/genética , Anopheles/metabolismo , Sistema Digestório/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Proteínas de Insetos/genética , Análise de Sequência com Séries de Oligonucleotídeos , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Proteínas de Protozoários/genética
18.
Genetics ; 167(4): 1781-90, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15342516

RESUMO

We report successful conditional gene expression in the malaria vector, Anopheles stephensi, on the basis of binary systems consisting of gene driver and responder transgenic lines generated by Minos-mediated germline transformation. An A. gambiae tissue-specific enhancer derived from a serpin (SRPN10) gene was utilized to control the temporal and spatial expression of doxycycline (dox)-sensitive transcriptional regulators in the driver lines. The "Tet-Off" driver utilized the tetracycline-controlled transcriptional activator (tTA) that is unable to bind and activate transcription from tetracycline operators (TetO) in the presence of dox; the "Tet-on" driver utilized the reverse tTA (rtTA) that, conversely, binds and activates TetO operators in the presence of dox. The responder lines carried insertions encompassing a LacZ reporter gene, cis-regulated by a TetO-P-element hybrid promoter. The progeny of crosses between driver and responder lines expressed beta-galactosidase under dual, tissue-specific and dox-mediated regulation. In adult rtTA/TetOPlacZ progeny, dox treatment rapidly induced beta-galactosidase activity throughout the midgut epithelium and especially in malaria parasite-invaded epithelial cells. Transactivator-dependent, dox-mediated regulation was observed in hemocytes and pericardial cells using both systems. Conditional tissue-specific regulation is a powerful tool for analyzing gene function in mosquitoes and potentially for development of strategies to control disease transmission.


Assuntos
Anopheles/genética , Regulação da Expressão Gênica , Animais , Animais Geneticamente Modificados , Doxiciclina/farmacologia , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Proteínas de Fluorescência Verde/genética , Malária/transmissão , Plasmídeos , Regiões Promotoras Genéticas/genética , Proteínas Recombinantes de Fusão/metabolismo , beta-Galactosidase/genética
19.
Mech Dev ; 117(1-2): 265-8, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12204268

RESUMO

The Drosophila C(2)-H(2)-type zinc-finger transcription factor CF2 has been shown to regulate follicular cell fate determination during oogenesis. Here we show that CF2 is also expressed in the developing muscles of the embryo where it first appears at stage 12 at the time of skeletal myoblast fusion. Later it is expressed in all muscle lineages including skeletal, visceral and cardiac. Epistatic analysis showed that CF2 expression is dependent on the myogenic factor MEF2.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Drosophila/embriologia , Drosophila/genética , Músculos/embriologia , Músculos/metabolismo , Fatores de Transcrição/genética , Animais , Proteínas de Ligação a DNA/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genes de Insetos , Imuno-Histoquímica , Fatores de Transcrição MEF2 , Fatores de Regulação Miogênica , Fatores de Transcrição/metabolismo , Dedos de Zinco/genética
20.
G3 (Bethesda) ; 3(7): 1191-4, 2013 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-23708298

RESUMO

We report the imminent completion of a set of reference genome assemblies for 16 species of Anopheles mosquitoes. In addition to providing a generally useful resource for comparative genomic analyses, these genome sequences will greatly facilitate exploration of the capacity exhibited by some Anopheline mosquito species to serve as vectors for malaria parasites. A community analysis project will commence soon to perform a thorough comparative genomic investigation of these newly sequenced genomes. Completion of this project via the use of short next-generation sequence reads required innovation in both the bioinformatic and laboratory realms, and the resulting knowledge gained could prove useful for genome sequencing projects targeting other unconventional genomes.


Assuntos
Anopheles/genética , Evolução Biológica , Genoma de Inseto , Malária/genética , Animais , Sequência de Bases , Biologia Computacional , Genômica , Humanos , Insetos Vetores/genética , Malária/parasitologia , Malária/transmissão , Análise de Sequência de DNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA