Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Cell ; 170(1): 158-171.e8, 2017 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-28666117

RESUMEN

Autophagy degrades cytoplasmic components and is important for development and human health. Although autophagy is known to be influenced by systemic intercellular signals, the proteins that control autophagy are largely thought to function within individual cells. Here, we report that Drosophila macroglobulin complement-related (Mcr), a complement ortholog, plays an essential role during developmental cell death and inflammation by influencing autophagy in neighboring cells. This function of Mcr involves the immune receptor Draper, suggesting a relationship between autophagy and the control of inflammation. Interestingly, Mcr function in epithelial cells is required for macrophage autophagy and migration to epithelial wounds, a Draper-dependent process. This study reveals, unexpectedly, that complement-related from one cell regulates autophagy in neighboring cells via an ancient immune signaling program.


Asunto(s)
Autofagia , Proteínas del Sistema Complemento/inmunología , Drosophila melanogaster/crecimiento & desarrollo , Animales , Citocinas , Proteínas de Drosophila , Drosophila melanogaster/citología , Drosophila melanogaster/inmunología , Inflamación/inmunología , Larva/crecimiento & desarrollo , Larva/inmunología , Macrófagos/inmunología , Glándulas Salivales/citología , Glándulas Salivales/crecimiento & desarrollo , Glándulas Salivales/metabolismo , Serpinas
2.
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
3.
Immunol Rev ; 319(1): 27-44, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37589239

RESUMEN

The clearance of dead and dying cells, termed efferocytosis, is a rapid and efficient process and one that is critical for organismal health. The extraordinary speed and efficiency with which dead cells are detected and engulfed by immune cells within tissues presents a challenge to researchers who wish to unravel this fascinating process, since these fleeting moments of uptake are almost impossible to catch in vivo. In recent years, the fruit fly (Drosophila melanogaster) embryo has emerged as a powerful model to circumvent this problem. With its abundance of dying cells, specialist phagocytes and relative ease of live imaging, the humble fly embryo provides a unique opportunity to catch and study the moment of cell engulfment in real-time within a living animal. In this review, we explore the recent advances that have come from studies in the fly, and how live imaging and genetics have revealed a previously unappreciated level of diversity in the efferocytic program. A variety of efferocytic strategies across the phagocytic cell population ensure efficient and rapid clearance of corpses wherever death is encountered within the varied and complex setting of a multicellular living organism.


Asunto(s)
Apoptosis , Drosophila melanogaster , Animales , Humanos , Fagocitosis , Fagocitos , Drosophila
4.
J Am Chem Soc ; 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38592946

RESUMEN

Selectively labeling cells with damaged membranes is needed not only for identifying dead cells in culture, but also for imaging membrane barrier dysfunction in pathologies in vivo. Most membrane permeability stains are permanently colored or fluorescent dyes that need washing to remove their non-uptaken extracellular background and reach good image contrast. Others are DNA-binding environment-dependent fluorophores, which lack design modularity, have potential toxicity, and can only detect permeabilization of cell volumes containing a nucleus (i.e., cannot delineate damaged volumes in vivo nor image non-nucleated cell types or compartments). Here, we develop modular fluorogenic probes that reveal the whole cytosolic volume of damaged cells, with near-zero background fluorescence so that no washing is needed. We identify a specific disulfonated fluorogenic probe type that only enters cells with damaged membranes, then is enzymatically activated and marks them. The esterase probe MDG1 is a reliable tool to reveal live cells that have been permeabilized by biological, biochemical, or physical membrane damage, and it can be used in multicolor microscopy. We confirm the modularity of this approach by also adapting it for improved hydrolytic stability, as the redox probe MDG2. We conclude by showing the unique performance of MDG probes in revealing axonal membrane damage (which DNA fluorogens cannot achieve) and in discriminating damage on a cell-by-cell basis in embryos in vivo. The MDG design thus provides powerful modular tools for wash-free in vivo imaging of membrane damage, and indicates how designs may be adapted for selective delivery of drug cargoes to these damaged cells: offering an outlook from selective diagnosis toward therapy of membrane-compromised cells in disease.

5.
Semin Cell Dev Biol ; 100: 160-166, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31812445

RESUMEN

Several cell lineages migrate through the developing and adult tissues of our bodies utilising a variety of modes of motility to suit the different substrates and environments they encounter en route to their destinations. Here we describe a novel adhesion-independent mode of single cell locomotion utilised by Drosophila fat body cells - the equivalent of vertebrate adipocytes. Like their human counterpart, these large cells were previously presumed to be immotile. However, in the Drosophila pupa fat body cells appear to be motile and migrate in a directed way towards wounds by peristaltic swimming through the hemolymph. The propulsive force is generated from a wave of cortical actomyosin that travels rearwards along the length of the cell. We discuss how this swimming mode of motility overcomes the physical constraints of microscopic objects moving in fluids, how fat body cells switch on other "motility machinery" to plug the wound on arrival, and whether other cell lineages in Drosophila and other organisms may, under certain circumstances, also adopt swimming as an effective mode of migration.


Asunto(s)
Adipocitos/citología , Movimiento Celular , Drosophila melanogaster/citología , Modelos Biológicos , Adipocitos/metabolismo , Animales , Adhesión Celular , Drosophila melanogaster/metabolismo , Pupa/citología , Pupa/metabolismo
6.
Int J Mol Sci ; 23(3)2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-35163253

RESUMEN

Cancer growth represents a dysregulated imbalance between cell gain and cell loss, where the rate of proliferating mutant tumour cells exceeds the rate of those that die. Apoptosis, the most renowned form of programmed cell death, operates as a key physiological mechanism that limits cell population expansion, either to maintain tissue homeostasis or to remove potentially harmful cells, such as those that have sustained DNA damage. Paradoxically, high-grade cancers are generally associated with high constitutive levels of apoptosis. In cancer, cell-autonomous apoptosis constitutes a common tumour suppressor mechanism, a property which is exploited in cancer therapy. By contrast, limited apoptosis in the tumour-cell population also has the potential to promote cell survival and resistance to therapy by conditioning the tumour microenvironment (TME)-including phagocytes and viable tumour cells-and engendering pro-oncogenic effects. Notably, the constitutive apoptosis-mediated activation of cells of the innate immune system can help orchestrate a pro-oncogenic TME and may also effect evasion of cancer treatment. Here, we present an overview of the implications of cell death programmes in tumour biology, with particular focus on apoptosis as a process with "double-edged" consequences: on the one hand, being tumour suppressive through deletion of malignant or pre-malignant cells, while, on the other, being tumour progressive through stimulation of reparatory and regenerative responses in the TME.


Asunto(s)
Apoptosis/fisiología , Neoplasias/patología , Animales , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Humanos , Inmunidad Innata/fisiología , Fagocitos/fisiología , Microambiente Tumoral/fisiología
7.
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
8.
J Immunol ; 198(9): 3679-3689, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28363904

RESUMEN

H2O2 is an early danger cue required for innate immune cell recruitment to wounds. To date, little is known about whether H2O2 is required for the migration of human adaptive immune cells to sites of inflammation. However, oxidative stress is known to impair T cell activity, induce actin stiffness, and inhibit cell polarization. In this study, we show that low oxidative concentrations of H2O2 also impede chemokinesis and chemotaxis of previously activated human T cells to CXCL11, but not CXCL10 or CXCL12. We show that this deficiency in migration is due to a reduction in inflammatory chemokine receptor CXCR3 surface expression and cellular activation of lipid phosphatase SHIP-1. We demonstrate that H2O2 acts through an Src kinase to activate a negative regulator of PI3K signaling, SHIP-1 via phosphorylation, providing a molecular mechanism for H2O2-induced chemotaxis deficiency. We hypothesize that although H2O2 serves as an early recruitment trigger for innate immune cells, it appears to operate as an inhibitor of T lymphocyte immune adaptive responses that are not required until later in the repair process.


Asunto(s)
Movimiento Celular , Quimiocina CXCL11/metabolismo , Peróxido de Hidrógeno/farmacología , Terapia de Inmunosupresión , Linfocitos T/efectos de los fármacos , Actinas/metabolismo , Inmunidad Adaptativa , Adulto , Anciano , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Femenino , Humanos , Inmunidad Innata/efectos de los fármacos , Masculino , Persona de Mediana Edad , Estrés Oxidativo/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/metabolismo , Receptores CXCR3/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Adulto Joven , Familia-src Quinasas/metabolismo
9.
Development ; 141(9): 1814-20, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24718989

RESUMEN

Wound repair is a fundamental, conserved mechanism for maintaining tissue homeostasis and shares many parallels with embryonic morphogenesis. Small wounds in simple epithelia rapidly assemble a contractile actomyosin cable at their leading edge, as well as dynamic filopodia that finally knit the wound edges together. Most studies of wound re-epithelialisation have focused on the actin machineries that assemble in the leading edge of front row cells and that resemble the contractile mechanisms that drive morphogenetic episodes, including Drosophila dorsal closure, but, clearly, multiple cell rows back must also contribute for efficient repair of the wound. Here, we examine the role of cells back from the wound edge and show that they also stretch towards the wound and cells anterior-posterior to the wound edge rearrange their junctions with neighbours to drive cell intercalation events. This process in anterior-posterior cells is active and dependent on pulses of actomyosin that lead to ratcheted shrinkage of junctions; the actomyosin pulses are targeted to breaks in the cell polarity protein Par3 at cell vertices. Inhibiting actomyosin dynamics back from the leading edge prevents junction shrinkage and inhibits the wound edge from advancing. These events recapitulate cell rearrangements that occur during germband extension, in which intercalation events drive the elongation of tissues.


Asunto(s)
Forma de la Célula , Drosophila melanogaster/citología , Epitelio/patología , Morfogénesis , Cicatrización de Heridas , Animales , Polaridad Celular , Drosophila melanogaster/embriología , Embrión no Mamífero/citología , Epitelio/embriología , Uniones Intercelulares/metabolismo , Miosinas/metabolismo
10.
J Cell Sci ; 126(Pt 15): 3475-84, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23704353

RESUMEN

Throughout embryonic development, macrophages not only act as the first line of defence against infection but also help to sculpt organs and tissues of the embryo by removing dead cells and secreting extracellular matrix components. Key to their function is the ability of embryonic macrophages to migrate and disperse throughout the embryo. Despite these important developmental functions, little is known about the molecular mechanisms underlying embryonic macrophage migration in vivo. Integrins are key regulators of many of the adult macrophage responses, but their role in embryonic macrophages remains poorly characterized. Here, we have used Drosophila macrophages (haemocytes) as a model system to address the role of integrins during embryonic macrophage dispersal in vivo. We show that the main ßPS integrin, myospheroid, affects haemocyte migration in two ways; by shaping the three-dimensional environment in which haemocytes migrate and by regulating the migration of haemocytes themselves. Live imaging revealed a requirement for myospheroid within haemocytes to coordinate the microtubule and actin dynamics, and to enable haemocyte developmental dispersal, contact repulsion and inflammatory migration towards wounds.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila/citología , Cadenas beta de Integrinas/metabolismo , Animales , Movimiento Celular/fisiología , Drosophila/embriología , Macrófagos/citología
11.
EMBO Rep ; 13(12): 1040-2, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23154465

RESUMEN

The fourth EMBO conference on 'The Molecular and Cellular Basis of Regeneration and Repair', held in September 2012, brought together researchers from both the regeneration and wound-healing fields. The meeting spanned a wide range of research topics from basic science to clinical application, and a veritable melting pot of model organisms and approaches resulted in an excellent fourth conference in this series.


Asunto(s)
Transformación Celular Neoplásica , Sistema Nervioso/crecimiento & desarrollo , Regeneración , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Humanos , Neoplasias/mortalidad , Neoplasias/patología , Especies Reactivas de Oxígeno/metabolismo , Cicatrización de Heridas
12.
Nat Cell Biol ; 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918597

RESUMEN

Ferroptosis is a distinct form of necrotic cell death caused by overwhelming lipid peroxidation, and emerging evidence indicates a major contribution to organ damage in multiple pathologies. However, ferroptosis has not yet been visualized in vivo due to a lack of specific probes, which has severely limited the study of how the immune system interacts with ferroptotic cells and how this process contributes to inflammation. Consequently, whether ferroptosis has a physiological role has remained a key outstanding question. Here we identify a distinct, ferroptotic-like, necrotic cell death occurring in vivo during wounding of the Drosophila embryo using live imaging. We further demonstrate that macrophages rapidly engage these necrotic cells within the embryo but struggle to engulf them, leading to prolonged, frustrated phagocytosis and frequent corpse disintegration. Conversely, suppression of the ferroptotic programme during wounding delays macrophage recruitment to the injury site, pointing to conflicting roles for ferroptosis during inflammation in vivo.

13.
Drug Alcohol Rev ; 2024 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-39031451

RESUMEN

INTRODUCTION: The Sydney Medically Supervised Injecting Centre provides a safe, non-judgemental space where people can inject pre-obtained substances under the supervision of trained staff. This article describes an unusual incident occurring at the Medically Supervised Injecting Centre in January 2023. CASE PRESENTATION: Two regular male clients attending the Medically Supervised Injecting Centre injected a substance they believed to be cocaine. Both clients experienced adverse reactions; one was transported to hospital, while the other became extremely distressed and agitated. Paraphernalia sent for testing returned a result of tiletamine (a dissociative used in veterinary medicine) and no cocaine, 30 h after the incident. DISCUSSION AND CONCLUSIONS: Where substances are novel or unknown, adverse events are often unexpected and may be more difficult to prepare for. Substance-induced acute agitation can be alarming and hazardous for people consuming drugs and those around them and may pose challenges for staff. There is a substantial evidence base for the benefits of on-site drug analysis and drug checking in reducing harms related to drug use, and in enhancing drug market monitoring. This incident was successfully managed by Medically Supervised Injecting Centre and hospital staff, with no major consequence, however clinical management could have been improved using point of care drug testing.

14.
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
15.
Adv Exp Med Biol ; 710: 49-57, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22127885

RESUMEN

The outcome of any bacterial infection, whether it is clearance of the infecting pathogen, establishment of a persistent infection, or even death of the host, is as dependent on the host as on the pathogen (Finlay and Falkow 1989). To infect a susceptible host bacterial pathogens express virulence factors, which alter host cell physiology and allow the pathogen to establish a nutrient-rich niche for growth and avoid clearance by the host immune response. However survival within the host often results in tissue damage, which to some cases accounts for the disease-specific pathology. For many bacterial pathogens the principal determinants of virulence and elicitors of host tissue damage are soluble exotoxins, which allow bacteria to penetrate into deeper tissue or pass through a host epithelial or endothelial barrier. Therefore, exploring the complex interplay between host tissue and bacterial toxins can help us to understand infectious disease and define the contributions of the host immune system to bacterial virulence. In this chapter, we describe a new model, the Drosophila embryo, for addressing a fundamental issue in bacterial pathogenesis, the elucidation of the in vivo targets of bacterial toxins and the monitoring of the first moments of the infection process in real-time. To develop this model, we used the insect and emerging human pathogen Photorhabdus asymbiotica and more specifically we characterised the initial cross-talk between the secreted cytotoxin Mcf1 and the embryonic hemocytes. Mcf1 is a potent cytotoxin which has been detected in all Photorhabdus strains isolated so far, which can rapidly kill insects upon injection. Despite several in vitro tissue culture studies, the biology of Mcf1 in vivo is not well understood. Furthermore, despite the identification of many Photorhabdus toxins using recombinant expression in E. coli (Waterfield et al. 2008), very few studies address the molecular mechanism of action of these toxins in relation to specific immune responses in vivo in the insect model.


Asunto(s)
Toxinas Bacterianas/farmacología , Drosophila/efectos de los fármacos , Drosophila/embriología , Drosophila/microbiología , Photorhabdus/química , Photorhabdus/patogenicidad , Factores de Virulencia/farmacología , Animales , Modelos Animales de Enfermedad , Hemocitos/efectos de los fármacos , Humanos
16.
Curr Biol ; 32(16): 3584-3592.e4, 2022 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-35835122

RESUMEN

Wound healing entails a fine balance between re-epithelialization and inflammation1,2 so that the risk of infection is minimized, tissue architecture is restored without scarring, and the epithelium regains its ability to withstand mechanical forces. How the two events are orchestrated in vivo remains poorly understood, largely due to the experimental challenges of simultaneously addressing mechanical and molecular aspects of the damage response. Here, exploiting Drosophila's genetic tractability and live imaging potential, we uncover a dual role for Piezo-a mechanosensitive channel involved in calcium influx3-during re-epithelialization and inflammation following injury in vivo. We show that loss of Piezo leads to faster wound closure due to increased wound edge intercalation and exacerbated myosin cable heterogeneity. Moreover, we show that loss of Piezo leads to impaired inflammation due to lower epidermal calcium levels and, subsequently, insufficient damage-induced ROS production. Despite initially appearing beneficial, loss of Piezo is severely detrimental to the long-term effectiveness of repair. In fact, wounds inflicted on Piezo knockout embryos become a permanent point of weakness within the epithelium, leading to impaired barrier function and reduced ability of wounded embryos to survive. In summary, our study uncovers a role for Piezo in regulating epithelial cell dynamics and immune cell responsiveness during damage repair in vivo. We propose a model whereby Piezo acts as molecular brake during wound healing, slowing down closure to ensure activation of sustained inflammation and re-establishment of a fully functional epithelial barrier.


Asunto(s)
Calcio , Cicatrización de Heridas , Epidermis , Epitelio , Humanos , Inflamación , Cicatrización de Heridas/genética
17.
Science ; 375(6585): 1182-1187, 2022 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-35271315

RESUMEN

Apoptosis of cells and their subsequent removal through efferocytosis occurs in nearly all tissues during development, homeostasis, and disease. However, it has been difficult to track cell death and subsequent corpse removal in vivo. We developed a genetically encoded fluorescent reporter, CharON (Caspase and pH Activated Reporter, Fluorescence ON), that could track emerging apoptotic cells and their efferocytic clearance by phagocytes. Using Drosophila expressing CharON, we uncovered multiple qualitative and quantitative features of coordinated clearance of apoptotic corpses during embryonic development. When confronted with high rates of emerging apoptotic corpses, the macrophages displayed heterogeneity in engulfment behaviors, leading to some efferocytic macrophages carrying high corpse burden. Overburdened macrophages were compromised in clearing wound debris. These findings reveal known and unexpected features of apoptosis and macrophage efferocytosis in vivo.


Asunto(s)
Apoptosis , Rastreo Celular , Drosophila/embriología , Desarrollo Embrionario , Macrófagos/fisiología , Fagocitosis , Animales , Concentración de Iones de Hidrógeno
18.
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
19.
J Cell Biol ; 173(3): 405-16, 2006 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-16651377

RESUMEN

Drosophila melanogaster hemocytes are highly motile macrophage-like cells that undergo a stereotypic pattern of migration to populate the whole embryo by late embryogenesis. We demonstrate that the migratory patterns of hemocytes at the embryonic ventral midline are orchestrated by chemotactic signals from the PDGF/VEGF ligands Pvf2 and -3 and that these directed migrations occur independently of phosphoinositide 3-kinase (PI3K) signaling. In contrast, using both laser ablation and a novel wounding assay that allows localized treatment with inhibitory drugs, we show that PI3K is essential for hemocyte chemotaxis toward wounds and that Pvf signals and PDGF/VEGF receptor expression are not required for this rapid chemotactic response. Our results demonstrate that at least two separate mechanisms operate in D. melanogaster embryos to direct hemocyte migration and show that although PI3K is crucial for hemocytes to sense a chemotactic gradient from a wound, it is not required to sense the growth factor signals that coordinate their developmental migrations along the ventral midline during embryogenesis.


Asunto(s)
Quimiotaxis/fisiología , Drosophila melanogaster/fisiología , Hemocitos/fisiología , Actinas/antagonistas & inhibidores , Actinas/metabolismo , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Movimiento Celular/fisiología , Quimiotaxis/efectos de los fármacos , Quimiotaxis/genética , Cromonas/farmacología , Citocalasina D/farmacología , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Proteínas del Huevo/genética , Embrión no Mamífero/citología , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Inhibidores Enzimáticos/farmacología , Regulación del Desarrollo de la Expresión Génica , Hemocitos/citología , Hemocitos/efectos de los fármacos , Hibridación in Situ , Microscopía Fluorescente , Morfolinas/farmacología , Mutación , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , ARN Interferente Pequeño/genética , Proteínas Tirosina Quinasas Receptoras/genética , Tiazoles/farmacología , Tiazolidinas , Factores de Tiempo
20.
Curr Biol ; 31(4): 875-883.e5, 2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33296680

RESUMEN

Drosophila provides a powerful model in which to study inflammation in vivo, and previous studies have revealed many of the key signaling events critical for recruitment of immune cells to tissue damage. In the fly, wounding stimulates the rapid production of hydrogen peroxide (H2O2).1,2 This then acts as an activation signal by triggering a signaling pathway within responding macrophages by directly activating the Src family kinase (SFK) Src42A,3 which in turn phosphorylates the damage receptor Draper. Activated Draper then guides macrophages to the wound through the detection of an as-yet unidentified chemoattractant.3-5 Similar H2O2-activated signaling pathways are also critical for leukocyte recruitment following wounding in larval zebrafish,6-9 where H2O2 activates the SFK Lyn to drive neutrophil chemotaxis. In this study, we combine proteomics, live imaging, and genetics in the fly to identify a novel regulator of inflammation in vivo; the PTP-type phosphatase Pez. Pez is expressed in macrophages and is critical for their efficient migration to wounds. Pez functions within activated macrophages downstream of damage-induced H2O2 and operates, via its band 4.1 ezrin, radixin, and moesin (FERM) domain, together with Src42A and Draper to ensure effective inflammatory cell recruitment to wounds. We show that this key role is conserved in vertebrates, because "crispant" zebrafish larvae of the Draper ortholog (MEGF10) or the Pez ortholog (PTPN21) exhibit a failure in leukocyte recruitment to wounds. This study demonstrates evolutionary conservation of inflammatory signaling and identifies MEGF10 and PTPN21 as potential therapeutic targets for the treatment of inflammatory disorders.


Asunto(s)
Proteínas de Drosophila , Proteínas de la Membrana , Proteínas Tirosina Fosfatasas no Receptoras , Pez Cebra , Animales , Drosophila , Peróxido de Hidrógeno , Inflamación/genética , Larva , Proteínas Tirosina Fosfatasas , Proteínas Proto-Oncogénicas pp60(c-src) , Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA