Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
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.

2.
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.

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.
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
5.
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
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.
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
9.
J Cell Biol ; 219(11)2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32915229

RESUMEN

Macroautophagy (autophagy) targets cytoplasmic cargoes to the lysosome for degradation. Like all vesicle trafficking, autophagy relies on phosphoinositide identity, concentration, and localization to execute multiple steps in this catabolic process. Here, we screen for phosphoinositide phosphatases that influence autophagy in Drosophila and identify CG3530. CG3530 is homologous to the human MTMR6 subfamily of myotubularin-related 3-phosphatases, and therefore, we named it dMtmr6. dMtmr6, which is required for development and viability in Drosophila, functions as a regulator of autophagic flux in multiple Drosophila cell types. The MTMR6 family member MTMR8 has a similar function in autophagy of higher animal cells. Decreased dMtmr6 and MTMR8 function results in autophagic vesicle accumulation and influences endolysosomal homeostasis.


Asunto(s)
Autofagia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Endosomas/metabolismo , Lisosomas/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Endocitosis , Femenino , Masculino , Fagocitosis , Transporte de Proteínas , Proteínas Tirosina Fosfatasas no Receptoras/genética , Homología de Secuencia
10.
Cell Rep ; 31(8): 107692, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32460022

RESUMEN

Macrophages must not only be responsive to an array of different stimuli, such as infection and cellular damage, but also perform phagocytosis within the diverse and complex tissue environments found in vivo. This requires a high degree of morphological and therefore cytoskeletal plasticity. Here, we use the exceptional genetics and in vivo imaging of Drosophila embryos to study macrophage phagocytic versatility during apoptotic corpse clearance. We find that macrophage phagocytosis is highly robust, arising from their possession of two distinct modes of engulfment that utilize exclusive suites of actin-regulatory proteins. "Lamellipodial phagocytosis" is Arp2/3-complex-dependent and allows cells to migrate toward and envelop apoptotic corpses. Alternatively, Diaphanous and Ena drive filopodial phagocytosis to reach out and draw in debris. Macrophages switch to "filopodial phagocytosis" to overcome spatial constraint, providing the robust plasticity necessary to ensure that whatever obstacle they encounter in vivo, they fulfil their critical clearance function.


Asunto(s)
Actinas/metabolismo , Drosophila/metabolismo , Macrófagos/metabolismo , Fagocitos/metabolismo , Animales , Transducción de Señal
11.
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
12.
Artículo en Inglés | MEDLINE | ID: mdl-31501193

RESUMEN

Multicellular organisms are not created through cell proliferation alone. It is through cell death that an indefinite cellular mass is pared back to reveal its true form. Cells are also lost throughout life as part of homeostasis and through injury. This detritus represents a significant burden to the living organism and must be cleared, most notably through the use of specialized phagocytic cells. Our understanding of these phagocytes and how they engulf cell corpses has been greatly aided by studying the fruit fly, Drosophila melanogaster Here we review the contribution of Drosophila research to our understanding of how phagocytes respond to cell death. We focus on the best studied phagocytes in the fly: the glia of the central nervous system, the ovarian follicle cells, and the macrophage-like hemocytes. Each is explored in the context of the tissue they maintain as well as how they function during development and in response to injury.


Asunto(s)
Sistema Nervioso Central/metabolismo , Drosophila melanogaster/inmunología , Drosophila melanogaster/metabolismo , Neuroglía/metabolismo , Fagocitos/citología , Animales , Apoptosis , Astrocitos/citología , Muerte Celular , Proliferación Celular , Proteínas de Drosophila/metabolismo , Femenino , Hemocitos/citología , Homeostasis , Inflamación , Macrófagos/inmunología , Macrófagos/metabolismo , Folículo Ovárico/inmunología , Folículo Ovárico/metabolismo , Fagocitosis
13.
Nat Cell Biol ; 21(11): 1370-1381, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31685997

RESUMEN

Cell migration is hypothesized to involve a cycle of behaviours beginning with leading edge extension. However, recent evidence suggests that the leading edge may be dispensable for migration, raising the question of what actually controls cell directionality. Here, we exploit the embryonic migration of Drosophila macrophages to bridge the different temporal scales of the behaviours controlling motility. This approach reveals that edge fluctuations during random motility are not persistent and are weakly correlated with motion. In contrast, flow of the actin network behind the leading edge is highly persistent. Quantification of actin flow structure during migration reveals a stable organization and asymmetry in the cell-wide flowfield that strongly correlates with cell directionality. This organization is regulated by a gradient of actin network compression and destruction, which is controlled by myosin contraction and cofilin-mediated disassembly. It is this stable actin-flow polarity, which integrates rapid fluctuations of the leading edge, that controls inherent cellular persistence.


Asunto(s)
Actinas/genética , Movimiento Celular/genética , Drosophila melanogaster/embriología , Mecanotransducción Celular , Pez Cebra/embriología , Actinas/metabolismo , Animales , Polaridad Celular , Rastreo Celular , Cofilina 1/genética , Cofilina 1/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hemocitos/citología , Hemocitos/metabolismo , Queratinocitos/citología , Queratinocitos/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Miosinas/genética , Miosinas/metabolismo , Cultivo Primario de Células , Pez Cebra/genética , Pez Cebra/metabolismo , Proteína Fluorescente Roja
14.
Curr Biol ; 29(22): 3851-3862.e4, 2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31668626

RESUMEN

In healthy individuals, injured tissues rapidly repair themselves following damage. Within a healing skin wound, recruited inflammatory cells release a multitude of bacteriocidal factors, including reactive oxygen species (ROS), to eliminate invading pathogens. Paradoxically, while these highly reactive ROS confer resistance to infection, they are also toxic to host tissues and may ultimately delay repair. Repairing tissues have therefore evolved powerful cytoprotective "resilience" machinery to protect against and tolerate this collateral damage. Here, we use in vivo time-lapse imaging and genetic manipulation in Drosophila to dissect the molecular and cellular mechanisms that drive tissue resilience to wound-induced stress. We identify a dynamic, cross-regulatory network of stress-activated cytoprotective pathways, linking calcium, JNK, Nrf2, and Gadd45, that act to both "shield" tissues from oxidative damage and promote efficient damage repair. Ectopic activation of these pathways confers stress protection to naive tissue, while their inhibition leads to marked delays in wound closure. Strikingly, the induction of cytoprotection is tightly linked to the pathways that initiate the inflammatory response, suggesting evolution of a fail-safe mechanism for tissue protection each time inflammation is triggered. A better understanding of these resilience mechanisms-their identities and precise spatiotemporal regulation-is of major clinical importance for development of therapeutic interventions for all pathologies linked to oxidative stress, including debilitating chronic non-healing wounds.


Asunto(s)
Citoprotección/fisiología , Estrés Oxidativo/fisiología , Cicatrización de Heridas/fisiología , Animales , Calcio/metabolismo , Citoprotección/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Inflamación/patología , Péptidos y Proteínas de Señalización Intracelular , Sistema de Señalización de MAP Quinasas , Factor 2 Relacionado con NF-E2 , Oxidación-Reducción , Estrés Oxidativo/genética , Especies Reactivas de Oxígeno/metabolismo , Cicatrización de Heridas/genética , Proteinas GADD45
15.
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
16.
Elife ; 72018 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-30260317

RESUMEN

Damage-associated molecular patterns (DAMPs) are molecules exposed or released by dead cells that trigger or modulate immunity and tissue repair. In vertebrates, the cytoskeletal component F-actin is a DAMP specifically recognised by DNGR-1, an innate immune receptor. Previously we suggested that actin is also a DAMP in Drosophila melanogaster by inducing STAT-dependent genes (Srinivasan et al., 2016). Here, we revise that conclusion and report that α-actinin is far more potent than actin at inducing the same STAT response and can be found in trace amounts in actin preparations. Recombinant expression of actin or α-actinin in bacteria demonstrated that only α-actinin could drive the expression of STAT target genes in Drosophila. The response to injected α-actinin required the same signalling cascade that we had identified in our previous work using actin preparations. Taken together, these data indicate that α-actinin rather than actin drives STAT activation when injected into Drosophila.


Asunto(s)
Actinina/farmacología , Actinas/farmacología , Proteínas de Drosophila/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Factores de Transcripción STAT/metabolismo , Actinina/administración & dosificación , Actinina/genética , Actinas/administración & dosificación , Actinas/genética , Animales , Animales Modificados Genéticamente , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
17.
J Vis Exp ; (136)2018 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-29985351

RESUMEN

During the rapid inflammatory response to tissue damage, cells of the innate immune system are quickly recruited to the injury site. Once at the wound, innate immune cells perform a number of essential functions, such as fighting infection, clearing necrotic debris, and stimulating matrix deposition. In order to fully understand the diverse signaling events that regulate this immune response, it is crucial to observe the complex behaviors of (and interactions that occur between) multiple cell lineages in vivo, and in real-time, with the high spatio-temporal resolution. The optical translucency and the genetic tractability of Drosophila embryos have established Drosophila as an invaluable model to live-image and dissect fundamental aspects of inflammatory cell behavior, including mechanisms of developmental dispersal, clearance of apoptotic corpses and/or microbial pathogens, and recruitment to wounds. However, more recent work has now demonstrated that employing a much later stage in the Drosophila lifecycle - the Drosophila pupa - offers a number of distinct advantages, including improved RNAi efficiency, longer imaging periods, and significantly greater immune cell numbers. Here we describe a protocol for imaging wound repair and the associated inflammatory response at the high spatio-temporal resolution in live Drosophila pupae. To follow the dynamics of both re-epithelialization and inflammation, we use a number of specific in vivo fluorescent markers for both the epithelium and innate immune cells. We also demonstrate the effectiveness of photo-convertible fluorophores, such as Kaede, for following the specific immune cell subsets, to track their behavior as they migrate to, and resolve from, the injury site.


Asunto(s)
Drosophila/fisiología , Inflamación/inmunología , Pupa/patogenicidad , Animales , Microscopía Confocal
18.
Dev Cell ; 44(4): 460-470.e3, 2018 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-29486196

RESUMEN

Adipocytes have many functions in various tissues beyond energy storage, including regulating metabolism, growth, and immunity. However, little is known about their role in wound healing. Here we use live imaging of fat body cells, the equivalent of vertebrate adipocytes in Drosophila, to investigate their potential behaviors and functions following skin wounding. We find that pupal fat body cells are not immotile, as previously presumed, but actively migrate to wounds using an unusual adhesion-independent, actomyosin-driven, peristaltic mode of motility. Once at the wound, fat body cells collaborate with hemocytes, Drosophila macrophages, to clear the wound of cell debris; they also tightly seal the epithelial wound gap and locally release antimicrobial peptides to fight wound infection. Thus, fat body cells are motile cells, enabling them to migrate to wounds to undertake several local functions needed to drive wound repair and prevent infections.


Asunto(s)
Adipocitos/metabolismo , Péptidos Catiónicos Antimicrobianos/farmacología , Movimiento Celular/fisiología , Drosophila melanogaster/fisiología , Infecciones por Escherichia coli/prevención & control , Cuerpo Adiposo/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Actomiosina/metabolismo , Adipocitos/citología , Animales , Células Cultivadas , Drosophila melanogaster/efectos de los fármacos , Escherichia coli , Infecciones por Escherichia coli/microbiología , Cuerpo Adiposo/citología
19.
Cell Rep ; 21(6): 1461-1470, 2017 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-29117553

RESUMEN

The most prominent developmental function attributed to the extracellular matrix (ECM) is cell migration. While cells in culture can produce ECM to migrate, the role of ECM in regulating developmental cell migration is classically viewed as an exogenous matrix presented to the moving cells. In contrast to this view, we show here that Drosophila embryonic hemocytes deposit their own laminins in streak-like structures to migrate efficiently throughout the embryo. With the help of transplantation experiments, live microscopy, and image quantification, we demonstrate that autocrine-produced laminin regulates hemocyte migration by controlling lamellipodia dynamics, stability, and persistence. Proper laminin deposition is regulated by the RabGTPase Rab8, which is highly expressed and required in hemocytes for lamellipodia dynamics and migration. Our results thus support a model in which, during embryogenesis, the Rab8-regulated autocrine deposition of laminin reinforces directional and effective migration by stabilizing cellular protrusions and strengthening otherwise transient adhesion states.


Asunto(s)
Proteínas de Drosophila/metabolismo , Laminina/metabolismo , Animales , Movimiento Celular , Células Cultivadas , Drosophila/crecimiento & desarrollo , Drosophila/metabolismo , Embrión no Mamífero/citología , Desarrollo Embrionario , Matriz Extracelular/metabolismo , GTP Fosfohidrolasas/metabolismo , Hemocitos/citología , Hemocitos/metabolismo , Microscopía Fluorescente , Seudópodos/fisiología
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...