Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
iScience ; 23(2): 100865, 2020 Feb 21.
Article in English | MEDLINE | ID: mdl-32058973

ABSTRACT

Clostridium difficile infections (CDIs) cause severe and occasionally life-threatening diarrhea. Hyper-virulent strains produce CDT, a toxin that ADP-ribosylates actin monomers and inhibits actin polymerization. We created transgenic Drosophila lines expressing the catalytic subunit CDTa to investigate its interaction with host signaling pathways in vivo. When expressed in the midgut, CDTa reduces body weight and fecal output and compromises survival, suggesting severe impairment of digestive functions. At the cellular level, CDTa induces F-actin network collapse, elimination of the intestinal brush border, and disruption of intercellular junctions. We confirm toxin-dependent re-distribution of Rab11 to enterocytes' apical surface and observe suppression of CDTa phenotypes by a Dominant-Negative form of Rab11 or RNAi of the dedicated Rab11GEF Crag (DENND4). We also report that Calmodulin (Cam) is required to mediate CDTa activity. In parallel, chemical inhibition of the Cam/Calcineurin pathway by Cyclosporin A or FK506 also reduces CDTa phenotypes, potentially opening new avenues for treating CDIs.

2.
Front Immunol ; 8: 1881, 2017.
Article in English | MEDLINE | ID: mdl-29326726

ABSTRACT

The swift clearance of apoptotic cells (ACs) (efferocytosis) by phagocytes is a critical event during development of all multicellular organisms. It is achieved through phagocytosis by professional or amateur phagocytes. Failure in this process can lead to the development of inflammatory autoimmune or neurodegenerative diseases. AC clearance has been conserved throughout evolution, although many details in its mechanisms remain to be explored. It has been studied in the context of mammalian macrophages, and in the nematode Caenorhabditis elegans, which lacks "professional" phagocytes such as macrophages, but in which other cell types can engulf apoptotic corpses. In Drosophila melanogaster, ACs are engulfed by macrophages, glial, and epithelial cells. Drosophila macrophages perform similar functions to those of mammalian macrophages. They are professional phagocytes that participate in phagocytosis of ACs and pathogens. Study of AC clearance in Drosophila has identified some key elements, like the receptors Croquemort and Draper, promoting Drosophila as a suitable model to genetically dissect this process. In this review, we survey recent works of AC clearance pathways in Drosophila, and discuss the physiological outcomes and consequences of this process.

3.
PLoS Pathog ; 12(10): e1005961, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27780230

ABSTRACT

Phagocytosis is an ancient mechanism central to both tissue homeostasis and immune defense. Both the identity of the receptors that mediate bacterial phagocytosis and the nature of the interactions between phagocytosis and other defense mechanisms remain elusive. Here, we report that Croquemort (Crq), a Drosophila member of the CD36 family of scavenger receptors, is required for microbial phagocytosis and efficient bacterial clearance. Flies mutant for crq are susceptible to environmental microbes during development and succumb to a variety of microbial infections as adults. Crq acts parallel to the Toll and Imd pathways to eliminate bacteria via phagocytosis. crq mutant flies exhibit enhanced and prolonged immune and cytokine induction accompanied by premature gut dysplasia and decreased lifespan. The chronic state of immune activation in crq mutant flies is further regulated by negative regulators of the Imd pathway. Altogether, our data demonstrate that Crq plays a key role in maintaining immune and organismal homeostasis.


Subject(s)
Drosophila Proteins/metabolism , Homeostasis , Immune System/immunology , Intestines/immunology , Intestines/microbiology , Phagocytosis/physiology , Receptors, Scavenger/metabolism , Aging , Animals , Drosophila Proteins/immunology , Drosophila melanogaster , Polymerase Chain Reaction , Receptors, Scavenger/immunology
4.
PLoS One ; 11(6): e0158217, 2016.
Article in English | MEDLINE | ID: mdl-27347682

ABSTRACT

Billions of cells die in our bodies on a daily basis and are engulfed by phagocytes. Engulfment, or phagocytosis, can be broken down into five basic steps: attraction of the phagocyte, recognition of the dying cell, internalization, phagosome maturation, and acidification. In this study, we focus on the last two steps, which can collectively be considered corpse processing, in which the engulfed material is degraded. We use the Drosophila ovarian follicle cells as a model for engulfment of apoptotic cells by epithelial cells. We show that engulfed material is processed using the canonical corpse processing pathway involving the small GTPases Rab5 and Rab7. The phagocytic receptor Draper is present on the phagocytic cup and on nascent, phosphatidylinositol 3-phosphate (PI(3)P)- and Rab7-positive phagosomes, whereas integrins are maintained on the cell surface during engulfment. Due to the difference in subcellular localization, we investigated the role of Draper, integrins, and downstream signaling components in corpse processing. We found that some proteins were required for internalization only, while others had defects in corpse processing as well. This suggests that several of the core engulfment proteins are required for distinct steps of engulfment. We also performed double mutant analysis and found that combined loss of draper and αPS3 still resulted in a small number of engulfed vesicles. Therefore, we investigated another known engulfment receptor, Crq. We found that loss of all three receptors did not inhibit engulfment any further, suggesting that Crq does not play a role in engulfment by the follicle cells. A more complete understanding of how the engulfment and corpse processing machinery interact may enable better understanding and treatment of diseases associated with defects in engulfment by epithelial cells.


Subject(s)
Phagocytes/physiology , Phagocytosis , Animals , Apoptosis , Caenorhabditis elegans , Drosophila , Endocytosis , Epithelial Cells/metabolism , Female , Integrins/metabolism , Membrane Proteins/metabolism , Mutation , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Phagosomes/metabolism , Transport Vesicles/metabolism
5.
Dev Cell ; 32(1): 19-30, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25533207

ABSTRACT

Clearance of apoptotic cells (efferocytosis) is achieved through phagocytosis by professional or amateur phagocytes. It is critical for tissue homeostasis and remodeling in all animals. Failure in this process can contribute to the development of inflammatory autoimmune or neurodegenerative diseases. We found previously that the PALL-SCF E3-ubiquitin ligase complex promotes apoptotic cell clearance, but it remained unclear how it did so. Here we show that the F-box protein PALL interacts with phosphorylated ribosomal protein S6 (RpS6) to promote its ubiquitylation and proteasomal degradation. This leads to RAC2 GTPase upregulation and activation and F-actin remodeling that promotes efferocytosis. We further show that the specific role of PALL in efferocytosis is driven by its apoptotic cell-induced nuclear export. Finding a role for RpS6 in the negative regulation of efferocytosis provides the opportunity to develop new strategies to regulate this process.


Subject(s)
Actin Cytoskeleton/physiology , Drosophila melanogaster/metabolism , Phagocytosis/physiology , Proteasome Endopeptidase Complex/metabolism , Ribosomal Protein S6/metabolism , Ubiquitin-Protein Ligases/metabolism , rac GTP-Binding Proteins/metabolism , Animals , Animals, Genetically Modified/growth & development , Animals, Genetically Modified/metabolism , Drosophila melanogaster/growth & development , Immunoprecipitation , Phosphorylation , Ribosomal Protein S6/genetics , Signal Transduction , Tandem Mass Spectrometry , Ubiquitin-Protein Ligases/genetics , Ubiquitination , rac GTP-Binding Proteins/genetics
6.
Neuron ; 81(3): 544-560, 2014 Feb 05.
Article in English | MEDLINE | ID: mdl-24412417

ABSTRACT

During developmental remodeling, neurites destined for pruning often degenerate on-site. Physical injury also induces degeneration of neurites distal to the injury site. Prompt clearance of degenerating neurites is important for maintaining tissue homeostasis and preventing inflammatory responses. Here we show that in both dendrite pruning and dendrite injury of Drosophila sensory neurons, epidermal cells rather than hemocytes are the primary phagocytes in clearing degenerating dendrites. Epidermal cells act via Draper-mediated recognition to facilitate dendrite degeneration and to engulf and degrade degenerating dendrites. Using multiple dendritic membrane markers to trace phagocytosis, we show that two members of the CD36 family, croquemort (crq) and debris buster (dsb), act at distinct stages of phagosome maturation for dendrite clearance. Our finding reveals the physiological importance of coordination between neurons and their surrounding epidermis, for both dendrite fragmentation and clearance.


Subject(s)
Dendrites/metabolism , Epidermal Cells , Epithelial Cells/physiology , Nerve Degeneration/physiopathology , Phagocytosis/physiology , Sensory Receptor Cells/cytology , Animals , Animals, Genetically Modified , CD36 Antigens/metabolism , Dendrites/ultrastructure , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , Larva , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Microscopy, Electron, Transmission , Mutation/genetics , Nerve Degeneration/pathology , Pupa , RNA Interference/physiology , Receptors, Scavenger/genetics , Receptors, Scavenger/metabolism , Time Factors
7.
PLoS One ; 7(2): e31488, 2012.
Article in English | MEDLINE | ID: mdl-22347485

ABSTRACT

Apoptosis, a genetically programmed cell death, allows for homeostasis and tissue remodelling during development of all multi-cellular organisms. Phagocytes swiftly recognize, engulf and digest apoptotic cells. Yet, to date the molecular mechanisms underlying this phagocytic process are still poorly understood. To delineate the molecular mechanisms of apoptotic cell clearance in Drosophila, we have carried out a deficiency screen and have identified three overlapping phagocytosis-defective mutants, which all delete the fly homologue of the ced-12 gene, known as Dmel\ced12. As anticipated, we have found that Dmel\ced-12 is required for apoptotic cell clearance, as for its C. elegans and mammalian homologues, ced-12 and elmo, respectively. However, the loss of Dmel\ced-12 did not solely account for the phenotypes of all three deficiencies, as zygotic mutations and germ line clones of Dmel\ced-12 exhibited weaker phenotypes. Using a nearby genetically interacting deficiency, we have found that the polycystic kidney disease 2 gene, pkd2, which encodes a member of the TRPP channel family, is also required for phagocytosis of apoptotic cells, thereby demonstrating a novel role for PKD2 in this process. We have also observed genetic interactions between pkd2, simu, drpr, rya-r44F, and retinophilin (rtp), also known as undertaker (uta), a gene encoding a MORN-repeat containing molecule, which we have recently found to be implicated in calcium homeostasis during phagocytosis. However, we have not found any genetic interaction between Dmel\ced-12 and simu. Based on these genetic interactions and recent reports demonstrating a role for the mammalian pkd-2 gene product in ER calcium release during store-operated calcium entry, we propose that PKD2 functions in the DRPR/RTP pathway to regulate calcium homeostasis during this process. Similarly to its C. elegans homologue, Dmel\Ced-12 appears to function in a genetically distinct pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/genetics , Drosophila Proteins/metabolism , Protein Kinases/metabolism , TRPP Cation Channels/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Calcium/metabolism , Drosophila , Drosophila Proteins/genetics , Mutation , Phagocytosis , Polycystic Kidney, Autosomal Dominant , Protein Kinase D2 , Protein Kinases/genetics , TRPP Cation Channels/genetics
8.
Cell ; 135(3): 524-34, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18984163

ABSTRACT

Phagocytosis is important during development and in the immune response for the removal of apoptotic cells and pathogens, yet its molecular mechanisms are poorly understood. In Caenorhabditis elegans, the CED2/5/10/12 pathway regulates actin during phagocytosis of apoptotic cells, whereas the role of the CED1/6/7 pathway in phagocytosis is unclear. We report that Undertaker (UTA), a Drosophila Junctophilin protein, is required for Draper (CED-1 homolog)-mediated phagocytosis. Junctophilins couple Ca2+ channels at the plasma membrane to those of the endoplasmic reticulum (ER), the Ryanodine receptors. We place Draper, its adaptor drCed-6, UTA, the Ryanodine receptor Rya-r44F, the ER Ca2+ sensor dSTIM, and the Ca2+-release-activated Ca2+ channel dOrai in the same pathway that promotes calcium homeostasis and phagocytosis. Thus, our results implicate a Junctophilin in phagocytosis and link Draper-mediated phagocytosis to Ca2+ homeostasis, highlighting a previously uncharacterized role for the CED1/6/7 pathway.


Subject(s)
Calcium/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/immunology , Membrane Proteins/metabolism , Phagocytosis , Animals , Animals, Genetically Modified , Apoptosis , Drosophila melanogaster/metabolism , Embryo, Nonmammalian , Eye Proteins
9.
Methods Enzymol ; 446: 39-59, 2008.
Article in English | MEDLINE | ID: mdl-18603115

ABSTRACT

Proper development of all multicellular organisms involves programmed apoptosis. Completion of this process requires removal of the resulting cell corpses through phagocytosis by their neighbors or by macrophages. Studies in C. elegans have been fruitful in the genetic dissection of key pathways, but they lack the professional immune system of higher organisms. Mammalian studies have identified a plethora of factors that are required for engulfment, but redundancy in the pathways has made it difficult to explain the genetic hierarchy of these factors. Thus, Drosophila has proven to be a useful evolutionary intermediate in which to examine this phenomenon. Here we describe methods used for dissecting the mechanisms and pathways involved in the engulfment of apoptotic cells by Drosophila phagocytes. Included are methods to be used for in vivo studies in the early embryo that can be used to examine engulfment of dying cells at various stages of embryogenesis. We also describe in vitro techniques for the use of Drosophila cell culture, including cell engulfment assays, that can be used for general phenotypic analysis, as well as live cell studies. We provide advice on imaging, including the preparation of samples for high-resolution microscopy and quantification of potential engulfment phenotypes for both in vivo and in vitro methods.


Subject(s)
Apoptosis/physiology , Drosophila/cytology , Phagocytosis/physiology , Acridine Orange , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line , Cell Separation/methods , Dactinomycin/analogs & derivatives , Dactinomycin/pharmacology , Drosophila/embryology , Embryo, Nonmammalian/cytology , Hemocytes/cytology , In Situ Nick-End Labeling/methods , Macrophages/cytology , Microscopy, Electron, Transmission/methods , Staining and Labeling/methods , Ultraviolet Rays
10.
Immunity ; 27(4): 585-96, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17936033

ABSTRACT

Many cells die by apoptosis during animal development. Apoptotic cells are rapidly removed through phagocytosis by their neighbors or by macrophages. To genetically dissect this process, we performed an in vivo screen for genes required for efficient phagocytosis of apoptotic cells by Drosophila macrophages and identified pallbearer (pall), which encodes an F box protein. F box proteins generally provide substrate specificity to Skp Cullin F box (SCF) complexes, acting as E3 ligases that target phosphorylated proteins to ubiquitylation and degradation via the 26S proteasome. We showed that Pallbearer functions in an SCF-dependent manner and provided direct evidence of a role for ubiquitylation and proteasomal degradation in phagocytosis of apoptotic corpses in vivo. This work might further our understanding of the regulation of apoptotic cell engulfment and thus our understanding of innate immunity as a whole.


Subject(s)
Apoptosis/physiology , Drosophila Proteins/metabolism , Macrophages/immunology , Phagocytosis/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Drosophila , Drosophila Proteins/genetics , Gene Expression , Immunohistochemistry , Immunoprecipitation , In Situ Hybridization , Macrophages/metabolism , Macrophages/ultrastructure , Microscopy, Confocal , Microscopy, Electron, Transmission , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/ultrastructure , Reverse Transcriptase Polymerase Chain Reaction , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Transfection , Ubiquitin-Protein Ligases/genetics
11.
Front Biosci ; 7: d1298-313, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11991836

ABSTRACT

Phagocytosis is the necessary corollary of apoptosis. It leads to the clearance of apoptotic cells by phagocytes, which can be 'professional' or 'amateur'. I review the known molecular aspects of phagocytosis of apoptotic corpses in mammals, Caenorhabditis elegans and Drosophila melanogaster from the point of view of the phagocyte and the apoptotic corpse. I highlight recent advances made in the field and discuss the physiological outcomes and consequences of this process. Indeed, phagocytosis of apoptotic cells is important in shaping or remodeling tissues to maintain their integrity and specialized functions during development and wound healing. It also contributes to the development of inflammation and/or its resolution after an injury or infection. This perhaps explains why the molecular mechanisms of phagocytosis of apoptotic cells are redundant and complex in mammals and suggests why they appear to have been mostly conserved through evolution. Caenorhabditis elegans has already proven to be useful in genetically dissecting the molecular mechanisms underlying phagocytosis of apoptotic corpses by 'amateur' neighboring cells. Drosophila melanogaster will become the model of choice in genetically dissecting the molecular mechanisms underlying phagocytosis of apoptotic cells by 'professional' phagocytes such as macrophages.


Subject(s)
Apoptosis/physiology , Caenorhabditis elegans/physiology , Drosophila melanogaster/physiology , Phagocytosis/physiology , Animals , Caenorhabditis elegans/cytology , Drosophila melanogaster/cytology , Humans , Macrophages/physiology , Models, Biological , Neutrophils/physiology , Receptors, Cell Surface/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...