Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 57(4): 613-631, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38599162

RESUMEN

While largely neglected over decades during which adaptive immunity captured most of the attention, innate immune mechanisms have now become central to our understanding of immunology. Innate immunity provides the first barrier to infection in vertebrates, and it is the sole mechanism of host defense in invertebrates and plants. Innate immunity also plays a critical role in maintaining homeostasis, shaping the microbiota, and in disease contexts such as cancer, neurodegeneration, metabolic syndromes, and aging. The emergence of the field of innate immunity has led to an expanded view of the immune system, which is no longer restricted to vertebrates and instead concerns all metazoans, plants, and even prokaryotes. The study of innate immunity has given rise to new concepts and language. Here, we review the history and definition of the core concepts of innate immunity, discussing their value and fruitfulness in the long run.


Asunto(s)
Inmunidad Innata , Memoria Inmunológica , Animales , Invertebrados , Inmunidad Adaptativa , Vertebrados
2.
Immunity ; 52(2): 374-387.e6, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32075729

RESUMEN

Animals require complex metabolic and physiological adaptations to maintain the function of vital organs in response to environmental stresses and infection. Here, we found that infection or injury in Drosophila induced the excretion of hemolymphatic lipids by Malpighian tubules, the insect kidney. This lipid purge was mediated by a stress-induced lipid-binding protein, Materazzi, which was enriched in Malpighian tubules. Flies lacking materazzi had higher hemolymph concentrations of reactive oxygen species (ROS) and increased lipid peroxidation. These flies also displayed Malpighian tubule dysfunction and were susceptible to infections and environmental stress. Feeding flies with antioxidants rescued the materazzi phenotype, indicating that the main role of Materazzi is to protect the organism from damage caused by stress-induced ROS. Our findings suggest that purging hemolymphatic lipids presents a physiological adaptation to protect host tissues from excessive ROS during immune and stress responses, a process that is likely to apply to other organisms.


Asunto(s)
Drosophila melanogaster/inmunología , Hemolinfa/metabolismo , Metabolismo de los Lípidos/inmunología , Túbulos de Malpighi/inmunología , Especies Reactivas de Oxígeno/inmunología , Inmunidad Adaptativa , Animales , Proteínas Portadoras/química , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Diglicéridos/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Heces/química , Peroxidación de Lípido/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Túbulos de Malpighi/metabolismo , Conformación Proteica , Especies Reactivas de Oxígeno/metabolismo , Estrés Fisiológico/inmunología
3.
Nat Immunol ; 17(10): 1150-8, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27548432

RESUMEN

The innate immune system needs to distinguish between harmful and innocuous stimuli to adapt its activation to the level of threat. How Drosophila mounts differential immune responses to dead and live Gram-negative bacteria using the single peptidoglycan receptor PGRP-LC is unknown. Here we describe rPGRP-LC, an alternative splice variant of PGRP-LC that selectively dampens immune response activation in response to dead bacteria. rPGRP-LC-deficient flies cannot resolve immune activation after Gram-negative infection and die prematurely. The alternative exon in the encoding gene, here called rPGRP-LC, encodes an adaptor module that targets rPGRP-LC to membrane microdomains and interacts with the negative regulator Pirk and the ubiquitin ligase DIAP2. We find that rPGRP-LC-mediated resolution of an efficient immune response requires degradation of activating and regulatory receptors via endosomal ESCRT sorting. We propose that rPGRP-LC selectively responds to peptidoglycans from dead bacteria to tailor the immune response to the level of threat.


Asunto(s)
Proteínas Portadoras/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Endosomas/metabolismo , Infecciones por Bacterias Gramnegativas/inmunología , Microdominios de Membrana/metabolismo , Pectobacterium carotovorum/inmunología , Isoformas de ARN/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Portadoras/metabolismo , Línea Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Exones/genética , Técnicas de Inactivación de Genes , Inmunidad/genética , Inmunomodulación , Proteínas Inhibidoras de la Apoptosis/metabolismo , Unión Proteica , Señales de Clasificación de Proteína/genética , Proteolisis , Isoformas de ARN/genética , Relación Estructura-Actividad
4.
Immunity ; 48(5): 897-910.e7, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29752064

RESUMEN

Intestinal infection triggers potent immune responses to combat pathogens and concomitantly drives epithelial renewal to maintain barrier integrity. Current models propose that epithelial renewal is primarily driven by damage caused by reactive oxygen species (ROS). Here we found that in Drosophila, the Imd-NF-κB pathway controlled enterocyte (EC) shedding upon infection, via a mechanism independent of ROS-associated apoptosis. Mechanistically, the Imd pathway synergized with JNK signaling to induce epithelial cell shedding specifically in the context of bacterial infection, requiring also the reduced expression of the transcription factor GATAe. Furthermore, cell-specific NF-κB responses enabled simultaneous production of antimicrobial peptides (AMPs) and epithelial shedding in different EC populations. Thus, the Imd-NF-κB pathway is central to the intestinal antibacterial response by mediating both AMP production and the maintenance of barrier integrity. Considering the similarities between Drosophila Imd signaling and mammalian TNFR pathway, our findings suggest the existence of an evolutionarily conserved genetic program in immunity-induced epithelial shedding.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/inmunología , Bacterias/inmunología , Infecciones Bacterianas/inmunología , Proteínas de Drosophila/inmunología , Células Epiteliales/inmunología , FN-kappa B/inmunología , Animales , Animales Modificados Genéticamente , Péptidos Catiónicos Antimicrobianos/metabolismo , Bacterias/crecimiento & desarrollo , Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/microbiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/inmunología , Drosophila melanogaster/metabolismo , Drosophila melanogaster/microbiología , Enterocitos/inmunología , Enterocitos/metabolismo , Enterocitos/microbiología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Factores de Transcripción GATA/genética , Factores de Transcripción GATA/inmunología , Factores de Transcripción GATA/metabolismo , Regulación de la Expresión Génica/inmunología , Mucosa Intestinal/citología , FN-kappa B/metabolismo , Transducción de Señal/inmunología
5.
Immunity ; 49(5): 929-942.e5, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30446385

RESUMEN

Commensal microbes colonize the gut epithelia of virtually all animals and provide several benefits to their hosts. Changes in commensal populations can lead to dysbiosis, which is associated with numerous pathologies and decreased lifespan. Peptidoglycan recognition proteins (PGRPs) are important regulators of the commensal microbiota and intestinal homeostasis. Here, we found that a null mutation in Drosophila PGRP-SD was associated with overgrowth of Lactobacillus plantarum in the fly gut and a shortened lifespan. L. plantarum-derived lactic acid triggered the activation of the intestinal NADPH oxidase Nox and the generation of reactive oxygen species (ROS). In turn, ROS production promoted intestinal damage, increased proliferation of intestinal stem cells, and dysplasia. Nox-mediated ROS production required lactate oxidation by the host intestinal lactate dehydrogenase, revealing a host-commensal metabolic crosstalk that is probably broadly conserved. Our findings outline a mechanism whereby host immune dysfunction leads to commensal dysbiosis that in turn promotes age-related pathologies.


Asunto(s)
Drosophila/fisiología , Ácido Láctico/metabolismo , Longevidad , Microbiota , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Disbiosis , Expresión Génica , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , L-Lactato Deshidrogenasa/genética , L-Lactato Deshidrogenasa/metabolismo , Mutación , NADPH Oxidasas/genética , Transducción de Señal , Simbiosis
6.
EMBO J ; 40(4): e104347, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33372708

RESUMEN

Adult stem cells must continuously fine-tune their behavior to regenerate damaged organs and avoid tumors. While several signaling pathways are well known to regulate somatic stem cells, the underlying mechanisms remain largely unexplored. Here, we demonstrate a cell-intrinsic role for the OvoL family transcription factor, Shavenbaby (Svb), in balancing self-renewal and differentiation of Drosophila intestinal stem cells. We find that svb is a downstream target of Wnt and EGFR pathways, mediating their activity for stem cell survival and proliferation. This requires post-translational processing of Svb into a transcriptional activator, whose upregulation induces tumor-like stem cell hyperproliferation. In contrast, the unprocessed form of Svb acts as a repressor that imposes differentiation into enterocytes, and suppresses tumors induced by altered signaling. We show that the switch between Svb repressor and activator is triggered in response to systemic steroid hormone, which is produced by ovaries. Therefore, the Svb axis allows intrinsic integration of local signaling cues and inter-organ communication to adjust stem cell proliferation versus differentiation, suggesting a broad role of OvoL/Svb in adult and cancer stem cells.


Asunto(s)
Diferenciación Celular , Autorrenovación de las Células , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Intestinos/fisiología , Células Madre/citología , Esteroides/farmacología , Factores de Transcripción/metabolismo , Animales , Proteínas de Unión al ADN/genética , Drosophila , Proteínas de Drosophila/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Células Madre/metabolismo , Factores de Transcripción/genética
7.
Immunity ; 45(5): 1013-1023, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27851910

RESUMEN

Activation of the innate immune response in Metazoans is initiated through the recognition of microbes by host pattern-recognition receptors. In Drosophila, diaminopimelic acid (DAP)-containing peptidoglycan from Gram-negative bacteria is detected by the transmembrane receptor PGRP-LC and by the intracellular receptor PGRP-LE. Here, we show that PGRP-SD acted upstream of PGRP-LC as an extracellular receptor to enhance peptidoglycan-mediated activation of Imd signaling. Consistent with this, PGRP-SD mutants exhibited impaired activation of the Imd pathway and increased susceptibility to DAP-type bacteria. PGRP-SD enhanced the localization of peptidoglycans to the cell surface and hence promoted signaling. Moreover, PGRP-SD antagonized the action of PGRP-LB, an extracellular negative regulator, to fine-tune the intensity of the immune response. These data reveal that Drosophila PGRP-SD functions as an extracellular receptor similar to mammalian CD14 and demonstrate that, comparable to lipopolysaccharide sensing in mammals, Drosophila relies on both intra- and extracellular receptors for the detection of bacteria.


Asunto(s)
Proteínas Portadoras/inmunología , Proteínas de Drosophila/inmunología , Drosophila melanogaster/inmunología , Infecciones por Bacterias Gramnegativas/inmunología , Factores Reguladores Miogénicos/inmunología , Transducción de Señal/inmunología , Animales , Modelos Animales de Enfermedad , Inmunidad Innata/inmunología , Peptidoglicano/inmunología , Reacción en Cadena de la Polimerasa , Receptores de Reconocimiento de Patrones/inmunología
8.
PLoS Genet ; 18(6): e1010259, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35714143

RESUMEN

Antimicrobial peptides (AMPs) are host-encoded antibiotics that combat invading pathogens. These genes commonly encode multiple products as post-translationally cleaved polypeptides. Recent studies have highlighted roles for AMPs in neurological contexts suggesting functions for these defence molecules beyond infection. During our immune study characterizing the antimicrobial peptide gene Baramicin, we recovered multiple Baramicin paralogs in Drosophila melanogaster and other species, united by their N-terminal IM24 domain. Not all paralogs were immune-induced. Here, through careful dissection of the Baramicin family's evolutionary history, we find that paralogs lacking immune induction result from repeated events of duplication and subsequent truncation of the coding sequence from an immune-inducible ancestor. These truncations leave only the IM24 domain as the prominent gene product. Surprisingly, using mutation and targeted gene silencing we demonstrate that two such genes are adapted for function in neural contexts in D. melanogaster. We also show enrichment in the head for independent Baramicin genes in other species. The Baramicin evolutionary history reveals that the IM24 Baramicin domain is not strictly useful in an immune context. We thus provide a case study for how an AMP-encoding gene might play dual roles in both immune and non-immune processes via its multiple peptide products. As many AMP genes encode polypeptides, a full understanding of how immune effectors interact with the nervous system will require consideration of all their peptide products.


Asunto(s)
Péptidos Catiónicos Antimicrobianos , Drosophila melanogaster , Animales , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Antimicrobianos , Sistema Nervioso
9.
Proc Natl Acad Sci U S A ; 119(30): e2208461119, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35858432

RESUMEN

Insects frequently harbor endosymbionts, which are bacteria housed within host tissues. These associations are stably maintained over evolutionary timescales through vertical transmission of endosymbionts from host mothers to their offspring. Some endosymbionts manipulate host reproduction to facilitate spread within natural populations. Consequently, such infections have major impacts on insect physiology and evolution. However, technical hurdles have limited our understanding of the molecular mechanisms underlying such insect-endosymbiont interactions. Here, we investigate the nutritional interactions between endosymbiotic partners using the tractable insect Drosophila melanogaster and its natural endosymbiont Spiroplasma poulsonii. Using a combination of functional assays, metabolomics, and proteomics, we show that the abundance and amino acid composition of a single Spiroplasma membrane lectin, Spiralin B (SpiB), dictates the amino acid requirements of the endosymbiont and determines its proliferation within host tissues. Ectopically increasing SpiB levels in host tissues disrupts localization of endosymbionts in the fly egg chambers and decreases vertical transmission. We find that SpiB is likely to be required by the endosymbiont to enter host oocytes, which may explain the massive investment of S. poulsonii in SpiB synthesis. SpiB both permits vertical transmission of the symbiont and limits its growth in nutrient-limiting conditions for the host; therefore, a single protein plays a pivotal role in ensuring durability of the interaction in a variable environment.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa , Drosophila melanogaster , Interacciones Microbiota-Huesped , Spiroplasma , Simbiosis , Aminoácidos/metabolismo , Animales , Proteínas de la Membrana Bacteriana Externa/metabolismo , Drosophila melanogaster/microbiología , Drosophila melanogaster/fisiología , Spiroplasma/metabolismo
10.
Nature ; 557(7704): 252-255, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29720654

RESUMEN

Several lineages of symbiotic bacteria in insects selfishly manipulate host reproduction to spread in a population 1 , often by distorting host sex ratios. Spiroplasma poulsonii2,3 is a helical and motile, Gram-positive symbiotic bacterium that resides in a wide range of Drosophila species 4 . A notable feature of S. poulsonii is male killing, whereby the sons of infected female hosts are selectively killed during development1,2. Although male killing caused by S. poulsonii has been studied since the 1950s, its underlying mechanism is unknown. Here we identify an S. poulsonii protein, designated Spaid, whose expression induces male killing. Overexpression of Spaid in D. melanogaster kills males but not females, and induces massive apoptosis and neural defects, recapitulating the pathology observed in S. poulsonii-infected male embryos5-11. Our data suggest that Spaid targets the dosage compensation machinery on the male X chromosome to mediate its effects. Spaid contains ankyrin repeats and a deubiquitinase domain, which are required for its subcellular localization and activity. Moreover, we found a laboratory mutant strain of S. poulsonii with reduced male-killing ability and a large deletion in the spaid locus. Our study has uncovered a bacterial protein that affects host cellular machinery in a sex-specific way, which is likely to be the long-searched-for factor responsible for S. poulsonii-induced male killing.


Asunto(s)
Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Drosophila melanogaster/microbiología , Caracteres Sexuales , Razón de Masculinidad , Spiroplasma/fisiología , Spiroplasma/patogenicidad , Simbiosis , Animales , Compensación de Dosificación (Genética)/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Femenino , Masculino , Cromosoma X/genética
11.
Proteins ; 91(12): 1571-1599, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37493353

RESUMEN

We present an in-depth analysis of selected CASP15 targets, focusing on their biological and functional significance. The authors of the structures identify and discuss key protein features and evaluate how effectively these aspects were captured in the submitted predictions. While the overall ability to predict three-dimensional protein structures continues to impress, reproducing uncommon features not previously observed in experimental structures is still a challenge. Furthermore, instances with conformational flexibility and large multimeric complexes highlight the need for novel scoring strategies to better emphasize biologically relevant structural regions. Looking ahead, closer integration of computational and experimental techniques will play a key role in determining the next challenges to be unraveled in the field of structural molecular biology.


Asunto(s)
Biología Computacional , Proteínas , Conformación Proteica , Modelos Moleculares , Biología Computacional/métodos , Proteínas/química
12.
PLoS Pathog ; 17(8): e1009846, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34432851

RESUMEN

The fruit fly Drosophila melanogaster combats microbial infection by producing a battery of effector peptides that are secreted into the haemolymph. Technical difficulties prevented the investigation of these short effector genes until the recent advent of the CRISPR/CAS era. As a consequence, many putative immune effectors remain to be formally described, and exactly how each of these effectors contribute to survival is not well characterized. Here we describe a novel Drosophila antifungal peptide gene that we name Baramicin A. We show that BaraA encodes a precursor protein cleaved into multiple peptides via furin cleavage sites. BaraA is strongly immune-induced in the fat body downstream of the Toll pathway, but also exhibits expression in other tissues. Importantly, we show that flies lacking BaraA are viable but susceptible to the entomopathogenic fungus Beauveria bassiana. Consistent with BaraA being directly antimicrobial, overexpression of BaraA promotes resistance to fungi and the IM10-like peptides produced by BaraA synergistically inhibit growth of fungi in vitro when combined with a membrane-disrupting antifungal. Surprisingly, BaraA mutant males but not females display an erect wing phenotype upon infection. Here, we characterize a new antifungal immune effector downstream of Toll signalling, and show it is a key contributor to the Drosophila antimicrobial response.


Asunto(s)
Antifúngicos/farmacología , Beauveria/efectos de los fármacos , Proteínas de Drosophila/farmacología , Drosophila melanogaster/efectos de los fármacos , Micosis/tratamiento farmacológico , Péptidos/farmacología , Animales , Beauveria/crecimiento & desarrollo , Beauveria/inmunología , Drosophila melanogaster/genética , Drosophila melanogaster/inmunología , Drosophila melanogaster/microbiología , Femenino , Masculino , Micosis/inmunología , Micosis/microbiología
13.
EMBO Rep ; 22(9): e52262, 2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34370384

RESUMEN

Programmed cell death plays a fundamental role in development and tissue homeostasis. Professional and non-professional phagocytes achieve the proper recognition, uptake, and degradation of apoptotic cells, a process called efferocytosis. Failure in efferocytosis leads to autoimmune and neurodegenerative diseases. In Drosophila, two transmembrane proteins of the Nimrod family, Draper and SIMU, mediate the recognition and internalization of apoptotic corpses. Beyond this early step, little is known about how apoptotic cell degradation is regulated. Here, we study the function of a secreted member of the Nimrod family, NimB4, and reveal its crucial role in the clearance of apoptotic cells. We show that NimB4 is expressed by macrophages and glial cells, the two main types of phagocytes in Drosophila. Similar to draper mutants, NimB4 mutants accumulate apoptotic corpses during embryogenesis and in the larval brain. Our study points to the role of NimB4 in phagosome maturation, more specifically in the fusion between the phagosome and lysosomes. We propose that similar to bridging molecules, NimB4 binds to apoptotic corpses to engage a phagosome maturation program dedicated to efferocytosis.


Asunto(s)
Drosophila , Fagocitos , Animales , Apoptosis/genética , Cadáver , Drosophila/genética , Fagocitosis , Fagosomas
14.
Proc Natl Acad Sci U S A ; 117(13): 7317-7325, 2020 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-32188787

RESUMEN

Iron sequestration is a recognized innate immune mechanism against invading pathogens mediated by iron-binding proteins called transferrins. Despite many studies on antimicrobial activity of transferrins in vitro, their specific in vivo functions are poorly understood. Here we use Drosophila melanogaster as an in vivo model to investigate the role of transferrins in host defense. We find that systemic infections with a variety of pathogens trigger a hypoferremic response in flies, namely, iron withdrawal from the hemolymph and accumulation in the fat body. Notably, this hypoferremia to infection requires Drosophila nuclear factor κB (NF-κB) immune pathways, Toll and Imd, revealing that these pathways also mediate nutritional immunity in flies. Next, we show that the iron transporter Tsf1 is induced by infections downstream of the Toll and Imd pathways and is necessary for iron relocation from the hemolymph to the fat body. Consistent with elevated iron levels in the hemolymph, Tsf1 mutants exhibited increased susceptibility to Pseudomonas bacteria and Mucorales fungi, which could be rescued by chemical chelation of iron. Furthermore, using siderophore-deficient Pseudomonas aeruginosa, we discover that the siderophore pyoverdine is necessary for pathogenesis in wild-type flies, but it becomes dispensable in Tsf1 mutants due to excessive iron present in the hemolymph of these flies. As such, our study reveals that, similar to mammals, Drosophila uses iron limitation as an immune defense mechanism mediated by conserved iron-transporting proteins transferrins. Our in vivo work, together with accumulating in vitro studies, supports the immune role of insect transferrins against infections via an iron withholding strategy.


Asunto(s)
Proteínas de Drosophila/metabolismo , Hierro/metabolismo , Transferrina/metabolismo , Animales , Proteínas de Drosophila/inmunología , Drosophila melanogaster , Hemolinfa/inmunología , Hemolinfa/metabolismo , Inmunidad Innata , Hierro/inmunología , FN-kappa B/metabolismo , Pseudomonas aeruginosa/metabolismo , Sideróforos/metabolismo , Transferrina/inmunología
15.
Annu Rev Genet ; 47: 377-404, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24016187

RESUMEN

The digestive tract plays a central role in the digestion and absorption of nutrients. Far from being a passive tube, it provides the first line of defense against pathogens and maintains energy homeostasis by exchanging neuronal and endocrine signals with other organs. Historically neglected, the gut of the fruit fly Drosophila melanogaster has recently come to the forefront of Drosophila research. Areas as diverse as stem cell biology, neurobiology, metabolism, and immunity are benefitting from the ability to study the genetics of development, growth regulation, and physiology in the same organ. In this review, we summarize our knowledge of the Drosophila digestive tract, with an emphasis on the adult midgut and its functional underpinnings.


Asunto(s)
Sistema Digestivo/anatomía & histología , Drosophila melanogaster/anatomía & histología , Animales , Dieta , Digestión , Sistema Digestivo/inmunología , Sistema Digestivo/inervación , Sistema Digestivo/microbiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/inmunología , Drosophila melanogaster/fisiología , Metabolismo Energético , Sistema Nervioso Entérico/fisiología , Células Enteroendocrinas/fisiología , Células Epiteliales/citología , Hormonas Gastrointestinales/fisiología , Interacciones Huésped-Patógeno , Absorción Intestinal , Larva , Longevidad , Moco/fisiología
16.
Cell Microbiol ; 22(5): e13156, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31912942

RESUMEN

Spiroplasma poulsonii is a vertically transmitted endosymbiont of Drosophila melanogaster that causes male-killing, that is the death of infected male embryos during embryogenesis. Here, we report a natural variant of S. poulsonii that is efficiently vertically transmitted yet does not selectively kill males, but kills rather a subset of all embryos regardless of their sex, a phenotype we call 'blind-killing'. We show that the natural plasmid of S. poulsonii has an altered structure: Spaid, the gene coding for the male-killing toxin, is deleted in the blind-killing strain, confirming its function as a male-killing factor. Then we further investigate several hypotheses that could explain the sex-independent toxicity of this new strain on host embryos. As the second non-male-killing variant isolated from a male-killing original population, this new strain raises questions on how male-killing is maintained or lost in fly populations. As a natural knock-out of Spaid, which is unachievable yet by genetic engineering approaches, this variant also represents a valuable tool for further investigations on the male-killing mechanism.


Asunto(s)
Drosophila/microbiología , Infecciones por Bacterias Gramnegativas/microbiología , Infecciones por Bacterias Gramnegativas/mortalidad , Spiroplasma/genética , Spiroplasma/metabolismo , Animales , Proteínas Bacterianas/genética , Drosophila/embriología , Drosophila melanogaster , Femenino , Regulación Bacteriana de la Expresión Génica , Infecciones por Bacterias Gramnegativas/veterinaria , Masculino , Fenotipo , Transcriptoma
17.
Appl Environ Microbiol ; 86(14)2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32444468

RESUMEN

Insects are frequently infected by bacterial symbionts that greatly affect their physiology and ecology. Most of these endosymbionts are, however, barely tractable outside their native host, rendering functional genetics studies difficult or impossible. Spiroplasma poulsonii is a facultative bacterial endosymbiont of Drosophila melanogaster that manipulates the reproduction of its host by killing its male progeny at the embryonic stage. S. poulsonii, although a very fastidious bacterium, is closely related to pathogenic Spiroplasma species that are cultivable and genetically modifiable. In this work, we present the transformation of S. poulsonii with a plasmid bearing a fluorescence cassette, leveraging techniques adapted from those used to modify the pathogenic species Spiroplasma citri We demonstrate the feasibility of S. poulsonii transformation and discuss approaches for mutant selection and fly colonization, which are persisting hurdles that must be overcome to allow functional bacterial genetics studies of this endosymbiont in vivoIMPORTANCE Dozens of bacterial endosymbiont species have been described and estimated to infect about half of all insect species. However, only a few them are tractable in vitro, which hampers our understanding of the bacterial determinants of the host-symbiont interaction. Developing a transformation method for S. poulsonii is a major step toward genomic engineering of this symbiont, which will foster basic research on endosymbiosis. This could also open the way to practical uses of endosymbiont engineering through paratransgenesis of vector or pest insects.


Asunto(s)
Drosophila melanogaster/microbiología , Drosophila melanogaster/fisiología , Spiroplasma/genética , Simbiosis , Transformación Bacteriana , Animales , Femenino , Masculino , Reproducción
18.
Immunity ; 35(5): 770-9, 2011 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-22118526

RESUMEN

Peptidoglycan recognition proteins (PGRPs) are key regulators of insect immune responses. In addition to recognition PGRPs, which activate the Toll and Imd pathways, the Drosophila genome encodes six catalytic PGRPs with the capacity to scavenge peptidoglycan. We have performed a systematic analysis of catalytic PGRP function using deletions, separately and in combination. Our findings support the role of PGRP-LB as a negative regulator of the Imd pathway and brought to light a synergy of PGRP-SCs with PGRP-LB in the systemic response. Flies lacking all six catalytic PGRPs were still viable but exhibited deleterious immune responses to innocuous gut infections. Together with recent studies on mammalian PGRPs, our study uncovers a conserved role for PGRPs in gut homeostasis. Analysis of the immune phenotype of flies lacking all catalytic PGRPs and the Imd regulator Pirk reveals that the Imd-mediated immune response is highly constrained by the existence of multiple negative feedbacks.


Asunto(s)
Amidohidrolasas/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/inmunología , Drosophila/microbiología , Bacterias Gramnegativas/inmunología , Amidohidrolasas/genética , Animales , Animales Modificados Genéticamente , Proteínas Portadoras/genética , Drosophila/genética , Proteínas de Drosophila/genética , Eliminación de Gen , Homeostasis/inmunología , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Mutación , Factores Reguladores Miogénicos/metabolismo , Fenotipo , Transducción de Señal
19.
PLoS Genet ; 13(6): e1006854, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28662029

RESUMEN

The speed of stem cell differentiation has to be properly coupled with self-renewal, both under basal conditions for tissue maintenance and during regeneration for tissue repair. Using the Drosophila midgut model, we analyze at the cellular and molecular levels the differentiation program required for robust regeneration. We observe that the intestinal stem cell (ISC) and its differentiating daughter, the enteroblast (EB), form extended cell-cell contacts in regenerating intestines. The contact between progenitors is stabilized by cell adhesion molecules, and can be dynamically remodeled to elicit optimal juxtacrine Notch signaling to determine the speed of progenitor differentiation. Notably, increasing the adhesion property of progenitors by expressing Connectin is sufficient to induce rapid progenitor differentiation. We further demonstrate that JAK/STAT signaling, Sox21a and GATAe form a functional relay to orchestrate EB differentiation. Thus, our study provides new insights into the complex and sequential events that are required for rapid differentiation following stem cell division during tissue replenishment.


Asunto(s)
Diferenciación Celular/genética , Proteínas de Drosophila/genética , Factores de Transcripción GATA/genética , Intestinos/crecimiento & desarrollo , Regeneración/genética , Factores de Transcripción SOXB2/genética , Animales , Adhesión Celular/genética , Comunicación Celular/genética , Proliferación Celular/genética , Autorrenovación de las Células , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Intestinos/citología , Quinasas Janus/genética , Factores de Transcripción STAT/genética , Transducción de Señal , Células Madre/citología
20.
BMC Microbiol ; 19(1): 46, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30786854

RESUMEN

BACKGROUND: Insects frequently live in close relationship with symbiotic bacteria that carry out beneficial functions for their host, like protection against parasites and viruses. However, in some cases, the mutualistic nature of such associations is put into question because of detrimental phenotypes caused by the symbiont. One example is the association between the vertically transmitted facultative endosymbiont Spiroplasma poulsonii and its natural host Drosophila melanogaster. Whereas S. poulsonii protects its host against parasitoid wasps and nematodes by the action of toxins from the family of Ribosome Inactivating Proteins (RIPs), the presence of S. poulsonii has been reported to reduce host's life span and to kill male embryos by a toxin called Spaid. In this work, we investigate the harmful effects of Spiroplasma RIPs on Drosophila in the absence of parasite infection. RESULTS: We show that only two Spiroplasma RIPs (SpRIP1 and SpRIP2) among the five RIP genes encoded in the S. poulsonii genome are significantly expressed during the whole Drosophila life cycle. Heterologous expression of SpRIP1 and 2 in uninfected flies confirms their toxicity, as indicated by a reduction of Drosophila lifespan and hemocyte number. We also show that RIPs can cause the death of some embryos, including females. CONCLUSION: Our results indicate that RIPs released by S. poulsonii contribute to the reduction of host lifespan and embryo mortality. This suggests that SpRIPs may impact the insect-symbiont homeostasis beyond their protective function against parasites.


Asunto(s)
Toxinas Bacterianas/genética , Drosophila melanogaster/microbiología , Interacciones Microbiota-Huesped , Proteínas Inactivadoras de Ribosomas/genética , Spiroplasma/química , Simbiosis , Animales , Proteínas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Embrión no Mamífero/microbiología , Femenino , Hemocitos , Hemolinfa/microbiología , Longevidad , Masculino , Proteínas Inactivadoras de Ribosomas/metabolismo , Spiroplasma/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA