Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
PLoS Pathog ; 12(7): e1005746, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27414410

RESUMEN

Cellular immune responses require the generation and recruitment of diverse blood cell types that recognize and kill pathogens. In Drosophila melanogaster larvae, immune-inducible lamellocytes participate in recognizing and killing parasitoid wasp eggs. However, the sequence of events required for lamellocyte generation remains controversial. To study the cellular immune system, we developed a flow cytometry approach using in vivo reporters for lamellocytes as well as for plasmatocytes, the main hemocyte type in healthy larvae. We found that two different blood cell lineages, the plasmatocyte and lamellocyte lineages, contribute to the generation of lamellocytes in a demand-adapted hematopoietic process. Plasmatocytes transdifferentiate into lamellocyte-like cells in situ directly on the wasp egg. In parallel, a novel population of infection-induced cells, which we named lamelloblasts, appears in the circulation. Lamelloblasts proliferate vigorously and develop into the major class of circulating lamellocytes. Our data indicate that lamellocyte differentiation upon wasp parasitism is a plastic and dynamic process. Flow cytometry with in vivo hemocyte reporters can be used to study this phenomenon in detail.


Asunto(s)
Proliferación Celular , Transdiferenciación Celular/fisiología , Drosophila melanogaster/fisiología , Drosophila melanogaster/parasitología , Hematopoyesis/fisiología , Hemocitos/citología , Avispas , Animales , Linaje de la Célula , Citometría de Flujo/métodos , Inmunohistoquímica , Larva , Microscopía Confocal
2.
BMC Genomics ; 18(1): 207, 2017 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-28249569

RESUMEN

BACKGROUND: Organisms typically face infection by diverse pathogens, and hosts are thought to have developed specific responses to each type of pathogen they encounter. The advent of transcriptomics now makes it possible to test this hypothesis and compare host gene expression responses to multiple pathogens at a genome-wide scale. Here, we performed a meta-analysis of multiple published and new transcriptomes using a newly developed bioinformatics approach that filters genes based on their expression profile across datasets. Thereby, we identified common and unique molecular responses of a model host species, the honey bee (Apis mellifera), to its major pathogens and parasites: the Microsporidia Nosema apis and Nosema ceranae, RNA viruses, and the ectoparasitic mite Varroa destructor, which transmits viruses. RESULTS: We identified a common suite of genes and conserved molecular pathways that respond to all investigated pathogens, a result that suggests a commonality in response mechanisms to diverse pathogens. We found that genes differentially expressed after infection exhibit a higher evolutionary rate than non-differentially expressed genes. Using our new bioinformatics approach, we unveiled additional pathogen-specific responses of honey bees; we found that apoptosis appeared to be an important response following microsporidian infection, while genes from the immune signalling pathways, Toll and Imd, were differentially expressed after Varroa/virus infection. Finally, we applied our bioinformatics approach and generated a gene co-expression network to identify highly connected (hub) genes that may represent important mediators and regulators of anti-pathogen responses. CONCLUSIONS: Our meta-analysis generated a comprehensive overview of the host metabolic and other biological processes that mediate interactions between insects and their pathogens. We identified key host genes and pathways that respond to phylogenetically diverse pathogens, representing an important source for future functional studies as well as offering new routes to identify or generate pathogen resilient honey bee stocks. The statistical and bioinformatics approaches that were developed for this study are broadly applicable to synthesize information across transcriptomic datasets. These approaches will likely have utility in addressing a variety of biological questions.


Asunto(s)
Abejas/genética , Interacciones Huésped-Patógeno/genética , Animales , Abejas/microbiología , Abejas/parasitología , Abejas/virología , Bases de Datos Genéticas , Evolución Molecular , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Inmunidad Innata/genética , Anotación de Secuencia Molecular , Nosema/fisiología , Virus ARN/fisiología , Varroidae/fisiología
3.
PLoS Pathog ; 11(5): e1004895, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25965263

RESUMEN

The cellular immune response against parasitoid wasps in Drosophila involves the activation, mobilization, proliferation and differentiation of different blood cell types. Here, we have assessed the role of Edin (elevated during infection) in the immune response against the parasitoid wasp Leptopilina boulardi in Drosophila melanogaster larvae. The expression of edin was induced within hours after a wasp infection in larval fat bodies. Using tissue-specific RNAi, we show that Edin is an important determinant of the encapsulation response. Although edin expression in the fat body was required for the larvae to mount a normal encapsulation response, it was dispensable in hemocytes. Edin expression in the fat body was not required for lamellocyte differentiation, but it was needed for the increase in plasmatocyte numbers and for the release of sessile hemocytes into the hemolymph. We conclude that edin expression in the fat body affects the outcome of a wasp infection by regulating the increase of plasmatocyte numbers and the mobilization of sessile hemocytes in Drosophila larvae.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Cuerpo Adiposo/metabolismo , Interacciones Huésped-Parásitos , Regulación hacia Arriba , Avispas/inmunología , Animales , Animales Modificados Genéticamente , Biomarcadores/metabolismo , Cruzamientos Genéticos , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Drosophila melanogaster/inmunología , Drosophila melanogaster/parasitología , Cuerpo Adiposo/citología , Cuerpo Adiposo/inmunología , Técnicas de Silenciamiento del Gen , Genes Reporteros , Hematopoyesis Extramedular , Hemocitos/citología , Hemocitos/inmunología , Hemocitos/metabolismo , Hemolinfa/citología , Hemolinfa/inmunología , Hemolinfa/metabolismo , Inmunidad Innata , Cinética , Larva/citología , Larva/inmunología , Larva/metabolismo , Larva/parasitología , Óvulo/inmunología , Óvulo/fisiología , Recuento de Huevos de Parásitos , Interferencia de ARN , Proteínas Recombinantes de Fusión/metabolismo , Avispas/fisiología
4.
EMBO Rep ; 16(12): 1664-72, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26412855

RESUMEN

The role of JAK/STAT signaling in the cellular immune response of Drosophila is not well understood. Here, we show that parasitoid wasp infection activates JAK/STAT signaling in somatic muscles of the Drosophila larva, triggered by secretion of the cytokines Upd2 and Upd3 from circulating hemocytes. Deletion of upd2 or upd3, but not the related os (upd1) gene, reduced the cellular immune response, and suppression of the JAK/STAT pathway in muscle cells reduced the encapsulation of wasp eggs and the number of circulating lamellocyte effector cells. These results suggest that JAK/STAT signaling in muscles participates in a systemic immune defense against wasp infection.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/inmunología , Drosophila/parasitología , Inmunidad Celular , Quinasas Janus/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Animales , Citocinas/metabolismo , Drosophila/genética , Proteínas de Drosophila/genética , Hemocitos/inmunología , Interacciones Huésped-Parásitos , Inmunidad Innata , Janus Quinasa 1/metabolismo , Quinasas Janus/genética , Larva/genética , Larva/inmunología , Larva/parasitología , Músculos/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal/inmunología , Factores de Transcripción/metabolismo , Avispas
5.
PLoS Pathog ; 8(8): e1002872, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22916019

RESUMEN

RNA interference (RNAi) is a major antiviral pathway that shapes evolution of RNA viruses. We show here that Nora virus, a natural Drosophila pathogen, is both a target and suppressor of RNAi. We detected viral small RNAs with a signature of Dicer-2 dependent small interfering RNAs in Nora virus infected Drosophila. Furthermore, we demonstrate that the Nora virus VP1 protein contains RNAi suppressive activity in vitro and in vivo that enhances pathogenicity of recombinant Sindbis virus in an RNAi dependent manner. Nora virus VP1 and the viral suppressor of RNAi of Cricket paralysis virus (1A) antagonized Argonaute-2 (AGO2) Slicer activity of RNA induced silencing complexes pre-loaded with a methylated single-stranded guide strand. The convergent evolution of AGO2 suppression in two unrelated insect RNA viruses highlights the importance of AGO2 in antiviral defense.


Asunto(s)
Proteínas Argonautas/metabolismo , Proteínas de Drosophila/metabolismo , Evolución Molecular , Silenciador del Gen , Virus de Insectos/metabolismo , Virus ARN/metabolismo , Animales , Proteínas Argonautas/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Virus de Insectos/genética , Virus ARN/genética
6.
Cells ; 13(7)2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38607032

RESUMEN

Coevolution of hosts and their parasites has shaped heterogeneity of effector hemocyte types, providing immune defense reactions with variable effectiveness. In this work, we characterize hemocytes of Drosophila willistoni, a species that has evolved a cellular immune system with extensive variation and a high degree of plasticity. Monoclonal antibodies were raised and used in indirect immunofluorescence experiments to characterize hemocyte subpopulations, follow their functional features and differentiation. Pagocytosis and parasitization assays were used to determine the functional characteristics of hemocyte types. Samples were visualized using confocal and epifluorescence microscopy. We identified a new multinucleated giant hemocyte (MGH) type, which differentiates in the course of the cellular immune response to parasitoids. These cells differentiate in the circulation through nuclear division and cell fusion, and can also be derived from the central hematopoietic organ, the lymph gland. They have a binary function as they take up bacteria by phagocytosis and are involved in the encapsulation and elimination of the parasitoid. Here, we show that, in response to large foreign particles, such as parasitoids, MGHs differentiate, have a binary function and contribute to a highly effective cellular immune response, similar to the foreign body giant cells of vertebrates.


Asunto(s)
Drosophila , Parásitos , Animales , Diferenciación Celular , Fagocitosis , Inmunidad Celular
7.
Front Immunol ; 14: 1322381, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38187383

RESUMEN

Background: Insects have specialized cell types that participate in the elimination of parasites, for instance, the lamellocytes of the broadly studied species Drosophila melanogaster. Other drosophilids, such as Drosophila ananassae and the invasive Zaprionus indianus, have multinucleated giant hemocytes, a syncytium of blood cells that participate in the encapsulation of the eggs or larvae of parasitoid wasps. These cells can be formed by the fusion of hemocytes in circulation or originate from the lymph gland. Their ultrastructure highly resembles that of the mammalian megakaryocytes. Methods: Morphological, protein expressional, and functional features of blood cells were revealed using epifluorescence and confocal microscopy. The respective hemocyte subpopulations were identified using monoclonal antibodies in indirect immunofluorescence assays. Fluorescein isothiocyanate (FITC)-labeled Escherichia coli bacteria were used in phagocytosis tests. Gene expression analysis was performed following mRNA sequencing of blood cells. Results: D. ananassae and Z. indianus encapsulate foreign particles with the involvement of multinucleated giant hemocytes and mount a highly efficient immune response against parasitoid wasps. Morphological, protein expressional, and functional assays of Z. indianus blood cells suggested that these cells could be derived from large plasmatocytes, a unique cell type developing specifically after parasitoid wasp infection. Transcriptomic analysis of blood cells, isolated from naïve and wasp-infected Z. indianus larvae, revealed several differentially expressed genes involved in signal transduction, cell movements, encapsulation of foreign targets, energy production, and melanization, suggesting their role in the anti-parasitoid response. A large number of genes that encode proteins associated with coagulation and wound healing, such as phenoloxidase activity factor-like proteins, fibrinogen-related proteins, lectins, and proteins involved in the differentiation and function of platelets, were constitutively expressed. The remarkable ultrastructural similarities between giant hemocytes and mammalian megakaryocytes, and presence of platelets, and giant cell-derived anucleated fragments at wound sites hint at the involvement of this cell subpopulation in wound healing processes, in addition to participation in the encapsulation reaction. Conclusion: Our observations provide insights into the broad repertoire of blood cell functions required for efficient defense reactions to maintain the homeostasis of the organism. The analysis of the differentiation and function of multinucleated giant hemocytes gives an insight into the diversification of the immune mechanisms.


Asunto(s)
Hemocitos , Avispas , Animales , Drosophila melanogaster , Diferenciación Celular , Drosophila , Plaquetas , Mamíferos
9.
J Immunol ; 184(11): 6188-98, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20421637

RESUMEN

Because NF-kappaB signaling pathways are highly conserved in evolution, the fruit fly Drosophila melanogaster provides a good model to study these cascades. We carried out an RNA interference (RNAi)-based genome-wide in vitro reporter assay screen in Drosophila for components of NF-kappaB pathways. We analyzed 16,025 dsRNA-treatments and identified 10 novel NF-kappaB regulators. Of these, nine dsRNA-treatments affect primarily the Toll pathway. G protein-coupled receptor kinase (Gprk)2, CG15737/Toll pathway activation mediating protein, and u-shaped were required for normal Drosomycin response in vivo. Interaction studies revealed that Gprk2 interacts with the Drosophila IkappaB homolog Cactus, but is not required in Cactus degradation, indicating a novel mechanism for NF-kappaB regulation. Morpholino silencing of the zebrafish ortholog of Gprk2 in fish embryos caused impaired cytokine expression after Escherichia coli infection, indicating a conserved role in NF-kappaB signaling. Moreover, small interfering RNA silencing of the human ortholog GRK5 in HeLa cells impaired NF-kappaB reporter activity. Gprk2 RNAi flies are susceptible to infection with Enterococcus faecalis and Gprk2 RNAi rescues Toll(10b)-induced blood cell activation in Drosophila larvae in vivo. We conclude that Gprk2/GRK5 has an evolutionarily conserved role in regulating NF-kappaB signaling.


Asunto(s)
Proteínas de Drosophila/inmunología , Quinasa 2 del Receptor Acoplado a Proteína-G/inmunología , Quinasa 5 del Receptor Acoplado a Proteína-G/metabolismo , Inmunidad Innata , FN-kappa B/inmunología , Transducción de Señal/fisiología , Animales , Western Blotting , Drosophila , Proteínas de Drosophila/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Quinasa 5 del Receptor Acoplado a Proteína-G/inmunología , Bacterias Gramnegativas Quimiolitotróficas/inmunología , Bacterias Gramnegativas Quimiolitotróficas/metabolismo , Humanos , Inmunohistoquímica , Inmunoprecipitación , FN-kappa B/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Pez Cebra
10.
Proc Natl Acad Sci U S A ; 106(12): 4805-9, 2009 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-19261847

RESUMEN

The blood cells, or hemocytes, in Drosophila participate in the immune response through the production of antimicrobial peptides, the phagocytosis of bacteria, and the encapsulation of larger foreign particles such as parasitic eggs; these immune reactions are mediated by phylogenetically conserved mechanisms. The encapsulation reaction is analogous to the formation of granuloma in vertebrates, and is mediated by large specialized cells, the lamellocytes. The origin of the lamellocytes has not been formally established, although it has been suggested that they are derived from the lymph gland, which is generally considered to be the main hematopoietic organ in the Drosophila larva. However, it was recently observed that a subepidermal population of sessile blood cells is released into the circulation in response to a parasitoid wasp infection. We set out to analyze this phenomenon systematically. As a result, we define the sessile hemocytes as a novel hematopoietic compartment, and the main source of lamellocytes.


Asunto(s)
Drosophila melanogaster/anatomía & histología , Drosophila melanogaster/inmunología , Hematopoyesis , Hemocitos/citología , Animales , Recuento de Células , Diferenciación Celular , Separación Celular , Drosophila melanogaster/citología , Proteínas Fluorescentes Verdes/metabolismo , Hemocitos/trasplante , Inmunidad , Larva/citología , Larva/inmunología , Larva/parasitología , Fenotipo , Factores de Tiempo
11.
Elife ; 112022 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-35920811

RESUMEN

Hemocytes, similar to vertebrate blood cells, play important roles in insect development and immunity, but it is not well understood how they perform their tasks. New technology, in particular single-cell transcriptomic analysis in combination with Drosophila genetics, may now change this picture. This review aims to make sense of recently published data, focusing on Drosophila melanogaster and comparing to data from other drosophilids, the malaria mosquito, Anopheles gambiae, and the silkworm, Bombyx mori. Basically, the new data support the presence of a few major classes of hemocytes: (1) a highly heterogenous and plastic class of professional phagocytes with many functions, called plasmatocytes in Drosophila and granular cells in other insects. (2) A conserved class of cells that control melanin deposition around parasites and wounds, called crystal cells in D. melanogaster, and oenocytoids in other insects. (3) A new class of cells, the primocytes, so far only identified in D. melanogaster. They are related to cells of the so-called posterior signaling center of the larval hematopoietic organ, which controls the hematopoiesis of other hemocytes. (4) Different kinds of specialized cells, like the lamellocytes in D. melanogaster, for the encapsulation of parasites. These cells undergo rapid evolution, and the homology relationships between such cells in different insects are uncertain. Lists of genes expressed in the different hemocyte classes now provide a solid ground for further investigation of function.


Asunto(s)
Bombyx , Drosophila , Animales , Drosophila melanogaster/genética , Hematopoyesis/genética , Hemocitos , Insectos
12.
J Innate Immun ; 14(4): 335-354, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34864742

RESUMEN

Multinucleated giant hemocytes (MGHs) represent a novel type of blood cell in insects that participate in a highly efficient immune response against parasitoid wasps involving isolation and killing of the parasite. Previously, we showed that circulating MGHs have high motility and the interaction with the parasitoid rapidly triggers encapsulation. However, structural and molecular mechanisms behind these processes remained elusive. Here, we used detailed ultrastructural analysis and live cell imaging of MGHs to study encapsulation in Drosophila ananassae after parasitoid wasp infection. We found dynamic structural changes, mainly driven by the formation of diverse vesicular systems and newly developed complex intracytoplasmic membrane structures, and abundant generation of giant cell exosomes in MGHs. In addition, we used RNA sequencing to study the transcriptomic profile of MGHs and activated plasmatocytes 72 h after infection, as well as the uninduced blood cells. This revealed that differentiation of MGHs was accompanied by broad changes in gene expression. Consistent with the observed structural changes, transcripts related to vesicular function, cytoskeletal organization, and adhesion were enriched in MGHs. In addition, several orphan genes encoding for hemolysin-like proteins, pore-forming toxins of prokaryotic origin, were expressed at high level, which may be important for parasitoid elimination. Our results reveal coordinated molecular and structural changes in the course of MGH differentiation and parasitoid encapsulation, providing a mechanistic model for a powerful innate immune response.


Asunto(s)
Hemocitos , Avispas , Animales , Drosophila , Interacciones Huésped-Parásitos , Inmunidad Innata , Transcriptoma , Avispas/genética
13.
Curr Biol ; 17(1): R22-4, 2007 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-17208173

RESUMEN

A new genetic study has shown that the phagocytic ability of Drosophila blood cells, the hemocytes, may be important for the further induction of an antibacterial response in other tissues.


Asunto(s)
Presentación de Antígeno/fisiología , Drosophila/inmunología , Cuerpo Adiposo/fisiología , Hemocitos/fisiología , Fagocitosis/fisiología , Animales
14.
Curr Biol ; 17(7): 649-54, 2007 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-17363253

RESUMEN

The hemocytes, the blood cells of Drosophila, participate in the humoral and cellular immune defense reactions against microbes and parasites [1-8]. The plasmatocytes, one class of hemocytes, are phagocytically active and play an important role in immunity and development by removing microorganisms as well as apoptotic cells. On the surface of circulating and sessile plasmatocytes, we have now identified a protein, Nimrod C1 (NimC1), which is involved in the phagocytosis of bacteria. Suppression of NimC1 expression in plasmatocytes inhibited the phagocytosis of Staphylococcus aureus. Conversely, overexpression of NimC1 in S2 cells stimulated the phagocytosis of both S. aureus and Escherichia coli. NimC1 is a 90-100 kDa single-pass transmembrane protein with ten characteristic EGF-like repeats (NIM repeats). The nimC1 gene is part of a cluster of ten related nimrod genes at 34E on chromosome 2, and similar clusters of nimrod-like genes are conserved in other insects such as Anopheles and Apis. The Nimrod proteins are related to other putative phagocytosis receptors such as Eater and Draper from D. melanogaster and CED-1 from C. elegans. Together, they form a superfamily that also includes proteins that are encoded in the human genome.


Asunto(s)
Proteínas de Drosophila/inmunología , Drosophila/inmunología , Hemocitos/inmunología , Fagocitosis , Receptores Inmunológicos/inmunología , Secuencias de Aminoácidos , Animales , Drosophila/citología , Drosophila/microbiología , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Escherichia coli/inmunología , Hemocitos/citología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Receptores Inmunológicos/química , Receptores Inmunológicos/genética , Staphylococcus aureus/inmunología
15.
Sci Rep ; 10(1): 19675, 2020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-33184473

RESUMEN

Nora virus, a virus of Drosophila, encapsidates one of the largest single-stranded RNA virus genomes known. Its taxonomic affinity is uncertain as it has a picornavirus-like cassette of enzymes for virus replication, but the capsid structure was at the time for genome publication unknown. By solving the structure of the virus, and through sequence comparison, we clear up this taxonomic ambiguity in the invertebrate RNA virosphere. Despite the lack of detectable similarity in the amino acid sequences, the 2.7 Å resolution cryoEM map showed Nora virus to have T = 1 symmetry with the characteristic capsid protein ß-barrels found in all the viruses in the Picornavirales order. Strikingly, α-helical bundles formed from the extended C-termini of capsid protein VP4B and VP4C protrude from the capsid surface. They are similar to signalling molecule folds and implicated in virus entry. Unlike other viruses of Picornavirales, no intra-pentamer stabilizing annulus was seen, instead the intra-pentamer stability comes from the interaction of VP4C and VP4B N-termini. Finally, intertwining of the N-termini of two-fold symmetry-related VP4A capsid proteins and RNA, provides inter-pentamer stability. Based on its distinct structural elements and the genetic distance to other picorna-like viruses we propose that Nora virus, and a small group of related viruses, should have its own family within the order Picornavirales.


Asunto(s)
Cápside/ultraestructura , Picornaviridae/ultraestructura , Receptores Virales/metabolismo , Sitios de Unión , Evolución Biológica , Cápside/metabolismo , Cápside/fisiología , Microscopía por Crioelectrón , Modelos Moleculares , Filogenia , Picornaviridae/clasificación , Picornaviridae/fisiología , Estabilidad Proteica , ARN Viral/metabolismo
16.
Dev Cell ; 5(3): 360-1, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12967553

RESUMEN

The fruit fly, Drosophila melanogaster, has become a popular tool for studying immediate reactions to environmental hazards, such as the heat shock and innate immune responses. In mammals, protective responses to infections and other insults are coordinated by a complex network of cytokines that mediate cell-to-cell signaling. By contrast, the corresponding heat shock and innate immune responses in Drosophila have usually been regarded as cell-autonomous processes. However, in this issue of Developmental Cell, show that cytokines do play a role in mediating an acute phase response in this organism.


Asunto(s)
Citocinas/fisiología , Drosophila melanogaster/fisiología , Respuesta al Choque Térmico/fisiología , Inmunidad Innata/fisiología , Estrés Fisiológico/metabolismo , Animales , Regulación de la Expresión Génica , Proteínas de Choque Térmico/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología
17.
Mol Biol Evol ; 25(11): 2337-47, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18703524

RESUMEN

The recently identified Nimrod superfamily is characterized by the presence of a special type of EGF repeat, the NIM repeat, located right after a typical CCXGY/W amino acid motif. On the basis of structural features, nimrod genes can be divided into three types. The proteins encoded by Draper-type genes have an EMI domain at the N-terminal part and only one copy of the NIM motif, followed by a variable number of EGF-like repeats. The products of Nimrod B-type and Nimrod C-type genes (including the eater gene) have different kinds of N-terminal domains, and lack EGF-like repeats but contain a variable number of NIM repeats. Draper and Nimrod C-type (but not Nimrod B-type) proteins carry a transmembrane domain. Several members of the superfamily were claimed to function as receptors in phagocytosis and/or binding of bacteria, which indicates an important role in the cellular immunity and the elimination of apoptotic cells. In this paper, the evolution of the Nimrod superfamily is studied with various methods on the level of genes and repeats. A hypothesis is presented in which the NIM repeat, along with the EMI domain, emerged by structural reorganizations at the end of an EGF-like repeat chain, suggesting a mechanism for the formation of novel types of repeats. The analyses revealed diverse evolutionary patterns in the sequences containing multiple NIM repeats. Although in the Nimrod B and Nimrod C proteins show characteristics of independent evolution, many internal NIM repeats in Eater sequences seem to have undergone concerted evolution. An analysis of the nimrod genes has been performed using phylogenetic and other methods and an evolutionary scenario of the origin and diversification of the Nimrod superfamily is proposed. Our study presents an intriguing example how the evolution of multigene families may contribute to the complexity of the innate immune response.


Asunto(s)
Evolución Molecular , Genes de Insecto , Familia de Multigenes , Secuencias Repetitivas de Aminoácido , Secuencias de Aminoácidos , Animales , Anopheles/genética , Abejas/genética , Drosophila/genética , Filogenia , Alineación de Secuencia , Tribolium/genética
18.
J Invertebr Pathol ; 101(1): 29-33, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19236875

RESUMEN

The biology of the Drosophila viruses has not been intensely investigated. Here we have investigated the biology of the Nora virus, a persistent Drosophila virus. We find that injected Nora virus is able to replicate in the files, reaching a high titer that is maintained in the next generation. There is a remarkable variation in the viral loads of individual flies in persistently infected stocks; the titers can differ by three orders of magnitude. The Nora virus is mainly found in the intestine of infected flies, and the histology of these infected intestines show increased vacuolization. The virus is excreted in the feces and is horizontally transmitted. The Nora virus infection has a very mild effect on the longevity of the flies, and no significant effect on the number of eggs laid and the percent of eggs that develop to adults.


Asunto(s)
Drosophila melanogaster/virología , Heces/virología , Picornaviridae/fisiología , Abdomen/anatomía & histología , Abdomen/virología , Animales , Femenino , Intestinos/ultraestructura , Intestinos/virología , Carga Viral , Replicación Viral
20.
Insect Biochem Mol Biol ; 37(4): 356-62, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17368199

RESUMEN

The NFAT gene encodes the only homolog in Drosophila of the five human Nuclear Factors of Activated T-cells, NFAT1-5. Its rel homology domain is most similar to that of NFAT5, and like the latter it lacks conserved AP1 and calcineurin binding sites. Two promoters give rise to alternative transcripts that are ubiquitously expressed in several different tissues. We generated mutants for each transcript, as well as a mutant that lacks all functional NFAT expression. Only the null mutant generated a visible phenotype, indicating that the two transcripts are redundant. The mutants are sensitive to high salt diet and have enlarged anal pads in hypotonic solution, suggesting that NFAT, like mammalian NFAT5, is regulating the osmotic balance. A phylogenetic reconstruction puts the Drosophila gene near the root of the NFAT tree, indicating that regulation of tonicity may be an ancestral function of the NFAT family.


Asunto(s)
Adaptación Fisiológica , Drosophila/fisiología , Factores de Transcripción NFATC/fisiología , Cloruro de Sodio/metabolismo , Animales , Drosophila/genética , Drosophila/metabolismo , Calor , Larva/metabolismo , Melaninas/metabolismo , Mutación , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA