Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cell ; 165(7): 1658-1671, 2016 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-27212238

RESUMEN

Macrophages are multifunctional cells that perform diverse roles in health and disease. Emerging evidence has suggested that these innate immune cells might also be capable of developing immunological memory, a trait previously associated with the adaptive system alone. While recent studies have focused on the dramatic macrophage reprogramming that follows infection and protects against secondary microbial attack, can macrophages also develop memory in response to other cues? Here, we show that apoptotic corpse engulfment by Drosophila macrophages is an essential primer for their inflammatory response to tissue damage and infection in vivo. Priming is triggered via calcium-induced JNK signaling, which leads to upregulation of the damage receptor Draper, thus providing a molecular memory that allows the cell to rapidly respond to subsequent injury or infection. This remarkable plasticity and capacity for memory places macrophages as key therapeutic targets for treatment of inflammatory disorders.


Asunto(s)
Drosophila/inmunología , Memoria Inmunológica , Macrófagos/inmunología , Animales , Apoptosis , Drosophila/citología , Proteínas de Drosophila/metabolismo , Escherichia coli , Inmunidad Innata , Sistema de Señalización de MAP Quinasas , Proteínas de la Membrana/metabolismo , Fagocitosis
2.
Clin Genet ; 105(5): 470-487, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38420660

RESUMEN

ASXL3-related disorder, sometimes referred to as Bainbridge-Ropers syndrome, was first identified as a distinct neurodevelopmental disorder by Bainbridge et al. in 2013. Since then, there have been a number of case series and single case reports published worldwide. A comprehensive review of the literature was carried out. Abstracts were screened, relevant literature was analysed, and descriptions of common phenotypic features were quantified. ASXL3 variants were collated and categorised. Common phenotypic features comprised global developmental delay or intellectual disability (97%), feeding problems (76%), hypotonia (88%) and characteristic facial features (93%). The majority of genetic variants were de novo truncating variants in exon 11 or 12 of the ASXL3 gene. Several gaps in our knowledge of this disorder were identified, namely, underlying pathophysiology and disease mechanism, disease contribution of missense variants, relevance of variant location, prevalence and penetrance data. Clinical information is currently limited by patient numbers and lack of longitudinal data, which this review aims to address.


Asunto(s)
Anomalías Múltiples , Discapacidades del Desarrollo , Facies , Discapacidad Intelectual , Trastornos del Neurodesarrollo , Niño , Humanos , Discapacidades del Desarrollo/genética , Factores de Transcripción/genética , Fenotipo , Síndrome , Discapacidad Intelectual/genética , Proteínas Represoras/genética
3.
J Cell Sci ; 132(5)2019 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-30718364

RESUMEN

The actin cytoskeleton is the engine that powers the inflammatory chemotaxis of immune cells to sites of tissue damage or infection. Here, we combine genetics with live in vivo imaging to investigate how cytoskeletal rearrangements drive macrophage recruitment to wounds in Drosophila We find that the actin-regulatory protein Ena is a master regulator of lamellipodial dynamics in migrating macrophages, where it remodels the cytoskeleton to form linear filaments that can then be bundled together by the cross-linker Fascin (also known as Singed in flies). In contrast, the formin Dia generates rare, probing filopods for specialised functions that are not required for migration. The role of Ena in lamellipodial bundling is so fundamental that its overexpression increases bundling even in the absence of Fascin by marshalling the remaining cross-linking proteins to compensate. This reorganisation of the lamellipod generates cytoskeletal struts that push against the membrane to drive leading edge advancement and boost cell speed. Thus, Ena-mediated remodelling extracts the most from the cytoskeleton to power robust macrophage chemotaxis during their inflammatory recruitment to wounds.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/fisiología , Forminas/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Complejos Multiproteicos/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Portadoras/metabolismo , Quimiotaxis , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Forminas/genética , Macrófagos/patología , Proteínas de Microfilamentos/metabolismo , Unión Proteica , Seudópodos/patología , Cicatrización de Heridas
4.
PLoS Biol ; 14(3): e1002395, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27015288

RESUMEN

Translation of hundreds of small ORFs (smORFs) of less than 100 amino acids has recently been revealed in vertebrates and Drosophila. Some of these peptides have essential and conserved cellular functions. In Drosophila, we have predicted a particular smORF class encoding ~80 aa hydrophobic peptides, which may function in membranes and cell organelles. Here, we characterise hemotin, a gene encoding an 88aa transmembrane smORF peptide localised to early endosomes in Drosophila macrophages. hemotin regulates endosomal maturation during phagocytosis by repressing the cooperation of 14-3-3ζ with specific phosphatidylinositol (PI) enzymes. hemotin mutants accumulate undigested phagocytic material inside enlarged endo-lysosomes and as a result, hemotin mutants have reduced ability to fight bacteria, and hence, have severely reduced life span and resistance to infections. We identify Stannin, a peptide involved in organometallic toxicity, as the Hemotin functional homologue in vertebrates, showing that this novel regulator of phagocytic processing is widely conserved, emphasizing the significance of smORF peptides in cell biology and disease.


Asunto(s)
Proteínas de Drosophila/genética , Endosomas/metabolismo , Macrófagos/metabolismo , Neuropéptidos/genética , Sistemas de Lectura Abierta , Proteínas 14-3-3/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Datos de Secuencia Molecular , Fagocitosis , Homología de Secuencia de Aminoácido
5.
Development ; 137(10): 1625-33, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20392742

RESUMEN

During embryonic development, Drosophila macrophages (haemocytes) undergo a series of stereotypical migrations to disperse throughout the embryo. One major migratory route is along the ventral nerve cord (VNC), where haemocytes are required for the correct development of this tissue. We show, for the first time, that a reciprocal relationship exists between haemocytes and the VNC and that defects in nerve cord development prevent haemocyte migration along this structure. Using live imaging, we demonstrate that the axonal guidance cue Slit and its receptor Robo are both required for haemocyte migration, but signalling is not autonomously required in haemocytes. We show that the failure of haemocyte migration along the VNC in slit mutants is not due to a lack of chemotactic signals within this structure, but rather to a failure in its detachment from the overlying epithelium, creating a physical barrier to haemocyte migration. This block of haemocyte migration in turn disrupts the formation of the dorsoventral channels within the VNC, further highlighting the importance of haemocyte migration for correct neural development. This study illustrates the important role played by the three-dimensional environment in directing cell migration in vivo and reveals an intriguing interplay between the developing nervous system and the blood cells within the fly, demonstrating that their development is both closely coupled and interdependent.


Asunto(s)
Movimiento Celular/fisiología , Drosophila/embriología , Macrófagos/fisiología , Sistema Nervioso/embriología , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Movimiento Celular/genética , Drosophila/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Embrión no Mamífero , Desarrollo Embrionario/genética , Hemocitos/metabolismo , Hemocitos/fisiología , Macrófagos/metabolismo , Modelos Biológicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/fisiología , Sistema Nervioso/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteínas Roundabout
6.
Front Immunol ; 14: 1310117, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38283366

RESUMEN

In Drosophila blood, plasmatocytes of the haemocyte lineage represent the functional equivalent of vertebrate macrophages and have become an established in vivo model with which to study macrophage function and behaviour. However, the use of plasmatocytes as a macrophage model has been limited by a historical perspective that plasmatocytes represent a homogenous population of cells, in contrast to the high levels of heterogeneity of vertebrate macrophages. Recently, a number of groups have reported transcriptomic approaches which suggest the existence of plasmatocyte heterogeneity, while we identified enhancer elements that identify subpopulations of plasmatocytes which exhibit potentially pro-inflammatory behaviours, suggesting conservation of plasmatocyte heterogeneity in Drosophila. These plasmatocyte subpopulations exhibit enhanced responses to wounds and decreased rates of efferocytosis when compared to the overall plasmatocyte population. Interestingly, increasing the phagocytic requirement placed upon plasmatocytes is sufficient to decrease the size of these plasmatocyte subpopulations in the embryo. However, the mechanistic basis for this response was unclear. Here, we examine how plasmatocyte subpopulations are modulated by apoptotic cell clearance (efferocytosis) demands and associated signalling pathways. We show that loss of the phosphatidylserine receptor Simu prevents an increased phagocytic burden from modulating specific subpopulation cells, while blocking other apoptotic cell receptors revealed no such rescue. This suggests that Simu-dependent efferocytosis is specifically involved in determining fate of particular subpopulations. Supportive of our original finding, mutations in amo (the Drosophila homolog of PKD2), a calcium-permeable channel which operates downstream of Simu, phenocopy simu mutants. Furthermore, we show that Amo is involved in the acidification of the apoptotic cell-containing phagosomes, suggesting that this reduction in pH may be associated with macrophage reprogramming. Additionally, our results also identify Ecdysone receptor signalling, a pathway related to control of cell death during developmental transitions, as a controller of plasmatocyte subpopulation identity. Overall, these results identify fundamental pathways involved in the specification of plasmatocyte subpopulations and so further validate Drosophila plasmatocytes as a heterogeneous population of macrophage-like cells within this important developmental and immune model.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila/metabolismo , Drosophila melanogaster/genética , Eferocitosis , Macrófagos/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
7.
PLoS Pathog ; 5(7): e1000518, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19609447

RESUMEN

Drosophila embryos are well studied developmental microcosms that have been used extensively as models for early development and more recently wound repair. Here we extend this work by looking at embryos as model systems for following bacterial infection in real time. We examine the behaviour of injected pathogenic (Photorhabdus asymbiotica) and non-pathogenic (Escherichia coli) bacteria and their interaction with embryonic hemocytes using time-lapse confocal microscopy. We find that embryonic hemocytes both recognise and phagocytose injected wild type, non-pathogenic E. coli in a Dscam independent manner, proving that embryonic hemocytes are phagocytically competent. In contrast, injection of bacterial cells of the insect pathogen Photorhabdus leads to a rapid 'freezing' phenotype of the hemocytes associated with significant rearrangement of the actin cytoskeleton. This freezing phenotype can be phenocopied by either injection of the purified insecticidal toxin Makes Caterpillars Floppy 1 (Mcf1) or by recombinant E. coli expressing the mcf1 gene. Mcf1 mediated hemocyte freezing is shibire dependent, suggesting that endocytosis is required for Mcf1 toxicity and can be modulated by dominant negative or constitutively active Rac expression, suggesting early and unexpected effects of Mcf1 on the actin cytoskeleton. Together these data show how Drosophila embryos can be used to track bacterial infection in real time and how mutant analysis can be used to genetically dissect the effects of specific bacterial virulence factors.


Asunto(s)
Drosophila/embriología , Embrión no Mamífero/metabolismo , Embrión no Mamífero/microbiología , Infecciones por Enterobacteriaceae/microbiología , Animales , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/farmacología , Moléculas de Adhesión Celular/metabolismo , Proteínas de Drosophila/metabolismo , Embrión no Mamífero/citología , Embrión no Mamífero/efectos de los fármacos , Infecciones por Enterobacteriaceae/metabolismo , Escherichia coli/metabolismo , Hemocitos/metabolismo , Microscopía Fluorescente , Microscopía por Video , Modelos Animales , Photorhabdus/metabolismo , Proteínas de Unión al GTP rac/metabolismo
8.
Front Cell Dev Biol ; 9: 636024, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33898424

RESUMEN

How multifunctional cells such as macrophages interpret the different cues within their environment and undertake an appropriate response is a key question in developmental biology. Understanding how cues are prioritized is critical to answering this - both the clearance of apoptotic cells (efferocytosis) and the migration toward damaged tissue is dependent on macrophages being able to interpret and prioritize multiple chemoattractants, polarize, and then undertake an appropriate migratory response. Here, we investigate the role of Spitz, the cardinal Drosophila epidermal growth factor (EGF) ligand, in regulation of macrophage behavior in the developing fly embryo, using activated variants with differential diffusion properties. Our results show that misexpression of activated Spitz can impact macrophage polarity and lead to clustering of cells in a variant-specific manner, when expressed either in macrophages or the developing fly heart. Spitz can also alter macrophage distribution and perturb apoptotic cell clearance undertaken by these phagocytic cells without affecting the overall levels of apoptosis within the embryo. Expression of active Spitz, but not a membrane-bound variant, can also increase macrophage migration speeds and impair their inflammatory responses to injury. The fact that the presence of Spitz specifically undermines the recruitment of more distal cells to wound sites suggests that Spitz desensitizes macrophages to wounds or is able to compete for their attention where wound signals are weaker. Taken together these results suggest this molecule regulates macrophage migration and their ability to dispose of apoptotic cells. This work identifies a novel regulator of Drosophila macrophage function and provides insights into signal prioritization and integration in vivo. Given the importance of apoptotic cell clearance and inflammation in human disease, this work may help us to understand the role EGF ligands play in immune cell recruitment during development and at sites of disease pathology.

9.
Dis Model Mech ; 4(1): 126-34, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21045209

RESUMEN

It is seldom the primary tumour that proves fatal in cancer, with metastasis the fundamental pathological process for disease progression. Upregulation of Mena, a member of the evolutionarily conserved Ena/VASP family of actin cytoskeletal regulators, promotes metastasis and invasive motility of breast cancer cells in vivo. To complement in vitro studies of Ena/VASP function in fibroblasts, we manipulated levels of Ena, the Drosophila homologue of Mena, in migrating embryonic macrophages (haemocytes). Consistent with data from fibroblasts in vitro, Ena localises to regions of actin dynamics within migrating haemocytes, stimulates lamellipodial dynamics and positively regulates the number and length of filopodia. However, whereas Ena overexpression in fibroblasts reduces migration speeds, overexpressing Ena in haemocytes leads to a dramatic increase in migration speeds, more closely resembling the increased motility of breast cancer cells that overexpress Mena. We provide evidence that this key difference is due to spatial constraints imposed on cells within the three-dimensional environment of the embryo; this might explain how Mena can be used to promote aggressive migratory behaviour during cancer progression.


Asunto(s)
Movimiento Celular , Proteínas de Unión al ADN/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/embriología , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Macrófagos/citología , Actinas/metabolismo , Animales , Conducta Animal , Drosophila melanogaster/metabolismo , Hemocitos/citología , Hemocitos/metabolismo , Macrófagos/metabolismo , Transporte de Proteínas , Seudópodos/metabolismo
10.
J Vis Exp ; (36)2010 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-20154641

RESUMEN

Many studies address cell migration using in vitro methods, whereas the physiologically relevant environment is that of the organism itself. Here we present a protocol for the mounting of Drosophila melanogaster embryos and subsequent live imaging of fluorescently labeled hemocytes, the embryonic macrophages of this organism. Using the Gal4-uas system we drive the expression of a variety of genetically encoded, fluorescently tagged markers in hemocytes to follow their developmental dispersal throughout the embryo. Following collection of embryos at the desired stage of development, the outer chorion is removed and the embryos are then mounted in halocarbon oil between a hydrophobic, gas-permeable membrane and a glass coverslip for live imaging. In addition to gross migratory parameters such as speed and directionality, higher resolution imaging coupled with the use of fluorescent reporters of F-actin and microtubules can provide more detailed information concerning the dynamics of these cytoskeletal components.


Asunto(s)
Movimiento Celular/fisiología , Drosophila melanogaster/embriología , Hemocitos/citología , Animales , Drosophila melanogaster/citología , Colorantes Fluorescentes/química , Proteínas Fluorescentes Verdes/química , Hemocitos/química
11.
J Cell Sci ; 120(Pt 2): 289-98, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17179204

RESUMEN

Ena/VASP proteins negatively regulate cell motility and contribute to repulsion from several guidance cues; however, there is currently no evidence for a role downstream of Eph receptors. Eph receptors mediate repulsion from ephrins at sites of intercellular contact during several developmental migrations. For example, the expression of ephrin-Bs in posterior halves of somites restricts neural crest cell migration to the anterior halves. Here we show that ephrin-B2 destabilises neural crest cell lamellipodia when presented in a substrate-bound or soluble form. Our timelapse studies show that repulsive events are associated with the rearward collapse and subsequent loss of lamellipodia as membrane ruffles. We hypothesise that Ena/VASP proteins contribute to repulsion from ephrins by destabilising cellular protrusions and show that Ena/VASP-deficient fibroblasts exhibit reduced repulsion from both ephrin-A and ephrin-B stripes compared to wild-type controls. Moreover, when EphB4 and ephrin-B2 were expressed in neighbouring Swiss 3T3 fibroblasts, VASP and Mena co-accumulated with activated Eph receptors at protrusions formed by EphB4-expressing cells. Sequestration of Ena/VASP proteins away from the periphery of these cells inhibited Eph receptor internalisation, a process that facilitates repulsion. Our results suggest that Ena/VASP proteins regulate ephrin-induced Eph receptor signalling events, possibly by destabilising lamellipodial protrusions.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Efrinas/metabolismo , Células 3T3 , Animales , Línea Celular Transformada , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Efrina-A5/metabolismo , Efrina-B2/metabolismo , Efrina-B2/farmacología , Fibroblastos/metabolismo , Fluoresceína-5-Isotiocianato , Técnica del Anticuerpo Fluorescente Indirecta , Colorantes Fluorescentes , Proteínas Fluorescentes Verdes/metabolismo , Ligandos , Ratones , Ratones Noqueados , Microscopía por Video , Cresta Neural/citología , Cresta Neural/efectos de los fármacos , Cresta Neural/embriología , Cresta Neural/metabolismo , Faloidina , Seudópodos/efectos de los fármacos , Seudópodos/fisiología , Ratas , Ratas Sprague-Dawley , Receptores de la Familia Eph/metabolismo , Solubilidad
12.
Exp Cell Res ; 310(2): 303-10, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16137676

RESUMEN

Vav family proteins act as guanine nucleotide exchange factors for Rho family proteins, which are known to orchestrate cytoskeletal changes and cell migration in response to extracellular stimuli. Using mice deficient for Vav1, Vav2 and/or Vav3, overlapping and isoform-specific functions of the three Vav proteins have been described in various hematopoietic cell types, but their roles in regulating cell morphology and migration have not been studied in detail. To investigate whether Vav isoforms have redundant or unique functions in regulating adhesion and migration, we investigated the properties of Vav1-deficient and Vav2-deficient macrophages. Both Vav1-deficient and Vav2-deficient cells have a smaller adhesive area; yet, only Vav1-deficient cells have a reduced migration speed, which coincides with a lower level of microtubules. Vav2-deficient macrophages display a high level of constitutive membrane ruffling, but neither Vav1 nor Vav2 is required for colony stimulating factor-1-induced membrane ruffling and cell spreading. Our results suggest that the migration speed of macrophages is regulated independently of spread area or membrane ruffling and that Vav1 is selectively required to maintain a normal migration speed.


Asunto(s)
Movimiento Celular , Citoesqueleto/ultraestructura , Macrófagos/inmunología , Macrófagos/ultraestructura , Proteínas Proto-Oncogénicas c-vav/fisiología , Animales , Adhesión Celular , Membrana Celular/metabolismo , Células Cultivadas , Proteínas del Citoesqueleto/metabolismo , Ratones , Ratones Mutantes , Mutación , Proteínas Proto-Oncogénicas c-vav/genética , Tubulina (Proteína)/metabolismo , Tubulina (Proteína)/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA