Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell ; 166(2): 314-327, 2016 Jul 14.
Article in English | MEDLINE | ID: mdl-27345367

ABSTRACT

Antigen presentation is essential for establishing immune tolerance and for immune responses against infectious disease and cancer. Although antigen presentation can be mediated by autophagy, here we demonstrate a pathway for mitochondrial antigen presentation (MitAP) that relies on the generation and trafficking of mitochondrial-derived vesicles (MDVs) rather than on autophagy/mitophagy. We find that PINK1 and Parkin, two mitochondrial proteins linked to Parkinson's disease (PD), actively inhibit MDV formation and MitAP. In absence of PINK1 or Parkin, inflammatory conditions trigger MitAP in immune cells, both in vitro and in vivo. MitAP and the formation of MDVs require Rab9 and Sorting nexin 9, whose recruitment to mitochondria is inhibited by Parkin. The identification of PINK1 and Parkin as suppressors of an immune-response-eliciting pathway provoked by inflammation suggests new insights into PD pathology.


Subject(s)
Antigen Presentation , Mitochondria/immunology , Parkinson Disease/immunology , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Dendritic Cells/pathology , Disease Models, Animal , Inflammation/metabolism , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Parkinson Disease/pathology , Protein Kinases/genetics , Transport Vesicles/metabolism , Ubiquitin-Protein Ligases/genetics
2.
Sci Transl Med ; 7(318): 318ra200, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26676607

ABSTRACT

Autoantibodies to components of apoptotic cells, such as anti-perlecan antibodies, contribute to rejection in organ transplant recipients. However, mechanisms of immunization to apoptotic components remain largely uncharacterized. We used large-scale proteomics, with validation by electron microscopy and biochemical methods, to compare the protein profiles of apoptotic bodies and apoptotic exosome-like vesicles, smaller extracellular vesicles released by endothelial cells downstream of caspase-3 activation. We identified apoptotic exosome-like vesicles as a central trigger for production of anti-perlecan antibodies and acceleration of rejection. Unlike apoptotic bodies, apoptotic exosome-like vesicles triggered the production of anti-perlecan antibodies in naïve mice and enhanced anti-perlecan antibody production and allograft inflammation in mice transplanted with an MHC (major histocompatibility complex)-incompatible aortic graft. The 20S proteasome core was active within apoptotic exosome-like vesicles and controlled their immunogenic activity. Finally, we showed that proteasome activity in circulating exosome-like vesicles increased after vascular injury in mice. These findings open new avenues for predicting and controlling maladaptive humoral responses to apoptotic cell components that enhance the risk of rejection after transplantation.


Subject(s)
Acute Kidney Injury/enzymology , Aorta/transplantation , Apoptosis/immunology , Autoantibodies/biosynthesis , Cell-Derived Microparticles/enzymology , Exosomes/enzymology , Graft Rejection/enzymology , Ischemia/enzymology , Proteasome Endopeptidase Complex/metabolism , Acute Kidney Injury/immunology , Acute Kidney Injury/pathology , Allografts , Animals , Aorta/enzymology , Aorta/immunology , Aorta/pathology , Autoantibodies/immunology , Biomarkers/metabolism , Cell-Derived Microparticles/immunology , Cell-Derived Microparticles/pathology , Cells, Cultured , Disease Models, Animal , Exosomes/immunology , Exosomes/pathology , Graft Rejection/immunology , Graft Rejection/pathology , Heparan Sulfate Proteoglycans/immunology , Heparan Sulfate Proteoglycans/metabolism , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Immunity, Humoral , Ischemia/immunology , Ischemia/pathology , Kidney Tubules, Proximal/enzymology , Kidney Tubules, Proximal/immunology , Kidney Tubules, Proximal/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/pathology , Peptide Fragments/immunology , Peptide Fragments/metabolism , Proteasome Endopeptidase Complex/immunology , Proteomics/methods , Rats , Time Factors
3.
J Cell Biol ; 210(7): 1133-52, 2015 Sep 28.
Article in English | MEDLINE | ID: mdl-26416964

ABSTRACT

Autophagy is a key degradative pathway coordinated by external cues, including starvation, oxidative stress, or pathogen detection. Rare are the molecules known to contribute mechanistically to the regulation of autophagy and expressed specifically in particular environmental contexts or in distinct cell types. Here, we unravel the role of RUN and FYVE domain-containing protein 4 (RUFY4) as a positive molecular regulator of macroautophagy in primary dendritic cells (DCs). We show that exposure to interleukin-4 (IL-4) during DC differentiation enhances autophagy flux through mTORC1 regulation and RUFY4 induction, which in turn actively promote LC3 degradation, Syntaxin 17-positive autophagosome formation, and lysosome tethering. Enhanced autophagy boosts endogenous antigen presentation by MHC II and allows host control of Brucella abortus replication in IL-4-treated DCs and in RUFY4-expressing cells. RUFY4 is therefore the first molecule characterized to date that promotes autophagy and influences endosome dynamics in a subset of immune cells.


Subject(s)
Autophagy/immunology , Dendritic Cells/immunology , Interleukin-4/immunology , Intracellular Signaling Peptides and Proteins/immunology , Lysosomes/immunology , Animals , Autophagy/genetics , Brucella abortus/immunology , Dendritic Cells/cytology , Interleukin-4/genetics , Intracellular Signaling Peptides and Proteins/genetics , Lysosomes/genetics , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/immunology , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/immunology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology
4.
J Virol ; 87(7): 3990-7, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23365427

ABSTRACT

Macroautophagy is a cellular pathway that degrades intracellular pathogens and contributes to antigen presentation. Herpes simplex virus 1 (HSV-1) infection triggers both macroautophagy and an additional form of autophagy that uses the nuclear envelope as a source of membrane. The present study constitutes the first in-depth analysis of nuclear envelope-derived autophagy (NEDA). We established LC3a as a marker that allowed us to distinguish between NEDA and macroautophagy in both immunofluorescence and flow cytometry. NEDA was observed in many different cell types, indicating that it is a general response to HSV-1 infection. This autophagic pathway is known to depend on the viral protein γ34.5, which can inhibit macroautophagy via binding to beclin-1. Using mutant viruses, we were able to show that binding of beclin-1 by γ34.5 had no effect on NEDA, demonstrating that NEDA is regulated differently than macroautophagy. Instead, NEDA was triggered in response to γ34.5 binding to protein phosphatase 1α, an interaction used by the virus to prevent host cells from shutting off protein translation. NEDA was not triggered when late viral protein production was inhibited with acyclovir or hippuristanol, indicating that the accumulation of these proteins might stress infected cells. Interestingly, expression of the late viral protein gH was sufficient to rescue NEDA in the context of infection with a virus that otherwise does not support strong late viral protein expression. We argue that NEDA is a cellular stress response triggered late during HSV-1 infection and might compensate for the viral alteration of the macroautophagic response.


Subject(s)
Autophagy/physiology , Herpes Simplex/physiopathology , Herpesvirus 1, Human/physiology , Nuclear Envelope/physiology , Protein Biosynthesis/physiology , Animals , Biomarkers/metabolism , DNA Primers/genetics , Flow Cytometry , Fluorescent Antibody Technique , Herpesvirus 1, Human/ultrastructure , Immunoblotting , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microscopy, Electron , Microtubule-Associated Proteins/metabolism , Viral Proteins/metabolism
5.
Proteomics ; 13(7): 1108-20, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23436686

ABSTRACT

The stress status of the apoptotic cell can promote phenotypic changes that have important consequences on the immunogenicity of the dying cell. Autophagy is one of the biological processes activated in response to a stressful condition. It is an important mediator of intercellular communications, both by regulating the unconventional secretion of molecules, including interleukin 1ß, and by regulating the extracellular release of ATP from early stage apoptotic cells. Additionally, autophagic components can be released in a caspase-dependent manner by serum-starved human endothelial cells that have engaged apoptotic and autophagic processes. The nature and the components of the extracellular vesicles released by dying autophagic cells are not known. In this study, we have identified extracellular membrane vesicles that are released by human endothelial cells undergoing apoptosis and autophagy, and characterized their biochemical, ultrastructural, morphological properties as well as their proteome. These extracellular vesicles differ from classical apoptotic bodies because they do not contain nucleus components and are released independently of Rho-associated, coiled-coil containing protein kinase 1 activation. Instead, they are enriched with autophagosomes and mitochondria and convey various danger signals, including ATP, suggesting that they could be involved in the modulation of innate immunity.


Subject(s)
Autophagy , Exosomes/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Culture Media, Serum-Free/pharmacology , Enzyme Activation/drug effects , Exosomes/ultrastructure , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/ultrastructure , Humans , Necrosis , Phagosomes/drug effects , Phagosomes/metabolism , Phagosomes/ultrastructure , Proteomics , rho-Associated Kinases/metabolism
6.
PLoS One ; 7(7): e42199, 2012.
Article in English | MEDLINE | ID: mdl-22860081

ABSTRACT

BACKGROUND: Senescent red blood cells (RBC) are recognized, phagocytosed and cleared by tissue macrophages. During this erythrophagocytosis (EP), RBC are engulfed and processed in special compartments called erythrophagosomes. We previously described that following EP, heme is rapidly degraded through the catabolic activity of heme oxygenase (HO). Extracted heme iron is then either exported or stored by macrophages. However, the cellular localization of the early steps of heme processing and iron extraction during EP remains to be clearly defined. METHODOLOGY/PRINCIPAL FINDINGS: We took advantage of our previously described cellular model of EP, using bone marrow-derived macrophages (BMDM). The subcellular localization of both inducible and constitutive isoforms of HO (HO-1 and HO-2), of the divalent metal transporters (Nramp1, Nramp2/DMT1, Fpn), and of the recently identified heme transporter HRG-1, was followed by fluorescence and electron microscopy during the earliest steps of EP. We also looked at some ER [calnexin, glucose-6-phosphatase (G6Pase) activity] and lysosomes (Lamp1) markers during EP. In both quiescent and LPS-activated BMDM, Nramp1 and Lamp1 were shown to be strong markers of the erythrophagolysosomal membrane. HRG-1 was also recruited to the erythrophagosome. Furthermore, we observed calnexin labeling and G6Pase activity at the erythrophagosomal membrane, indicating the contribution of ER in this phagocytosis model. In contrast, Nramp2/DMT1, Fpn, HO-1 and HO-2 were not detected at the membrane of erythrophagosomes. CONCLUSIONS/SIGNIFICANCE: Our study highlights the subcellular localization of various heme- and iron-related proteins during early steps of EP, thereby suggesting a model for heme catabolism occurring outside the phagosome, with heme likely being transported into the cytosol through HRG1. The precise function of Nramp1 at the phagosomal membrane in this model remains to be determined.


Subject(s)
Erythrocytes/metabolism , Heme/metabolism , Iron/metabolism , Phagocytosis , Subcellular Fractions/metabolism , Animals , Cells, Cultured , Culture Media, Conditioned , Mice , Mice, Inbred Strains , Microscopy, Electron , Microscopy, Fluorescence
7.
Mol Cell Proteomics ; 11(7): M111.016378, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22427703

ABSTRACT

Phagosomes, by killing and degrading pathogens for antigen presentation, are organelles implicated in key aspects of innate and adaptive immunity. Although it has been well established that phagosomes consist of membranes from the plasma membrane, endosomes, and lysosomes, the notion that the endoplasmic reticulum (ER) membrane could play an important role in the formation of the phagosome is debated. However, a method to accurately estimate the contribution of potential source organelles and contaminants to the phagosome proteome has been lacking. Herein, we have developed a proteomic approach for objectively quantifying the contribution of various organelles to the early and late phagosomes by comparing these fractions to their total membrane and postnuclear supernatant of origin in the J774A.1 murine macrophage cell line. Using quantitative label-free mass spectrometry, the abundance of peptides corresponding to hundreds of proteins was estimated and attributed to one of five organelles (e.g. plasma membrane, endosomes/lysosomes, ER, Golgi, and mitochondria). These data in combination with a stable isotope labeling in cell culture method designed to detect potential contaminant sources revealed that the ER is part of the phagosomal membrane and contributes ≈ 20% of the early phagosome proteome. In addition, only a subset of ER proteins is recruited to the phagosome, suggesting that a specific subdomain(s) of the ER might be involved in phagocytosis. Western blotting and immunofluorescence substantially validated this conclusion; we were able to demonstrate that the fraction of the ER in which the ER marker GFP-KDEL accumulates is excluded from the phagosomes, whereas that containing the mVenus-Syntaxin 18 is recruited. These results highlight promising new avenues for the description of the pathogenic mechanisms used by Leishmania, Brucella, and Legionella spp., which thrive in ER-rich phagosomes.


Subject(s)
Endoplasmic Reticulum/chemistry , Macrophages/metabolism , Phagosomes/chemistry , Proteomics/methods , Animals , Biomarkers/analysis , Blotting, Western , Cell Line , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Endosomes/chemistry , Endosomes/metabolism , Endosomes/ultrastructure , Fluorescent Antibody Technique , Golgi Apparatus/chemistry , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Isotope Labeling , Lysosomes/chemistry , Lysosomes/metabolism , Lysosomes/ultrastructure , Macrophages/cytology , Macrophages/ultrastructure , Mass Spectrometry , Mice , Oligopeptides , Phagocytosis , Phagosomes/metabolism , Phagosomes/ultrastructure , Plasmids , Protein Sorting Signals , Qa-SNARE Proteins , Transfection
8.
Nat Immunol ; 10(5): 480-7, 2009 May.
Article in English | MEDLINE | ID: mdl-19305394

ABSTRACT

Viral proteins are usually processed by the 'classical' major histocompatibility complex (MHC) class I presentation pathway. Here we showed that although macrophages infected with herpes simplex virus type 1 (HSV-1) initially stimulated CD8(+) T cells by this pathway, a second pathway involving a vacuolar compartment was triggered later during infection. Morphological and functional analyses indicated that distinct forms of autophagy facilitated the presentation of HSV-1 antigens on MHC class I molecules. One form of autophagy involved a previously unknown type of autophagosome that originated from the nuclear envelope. Whereas interferon-gamma stimulated classical MHC class I presentation, fever-like hyperthermia and the pyrogenic cytokine interleukin 1beta activated autophagy and the vacuolar processing of viral peptides. Viral peptides in autophagosomes were further processed by the proteasome, which suggests a complex interaction between the vacuolar and MHC class I presentation pathways.


Subject(s)
Antigen Presentation/immunology , Autophagy/immunology , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Histocompatibility Antigens Class I/immunology , Animals , Antigens, Viral/immunology , Cytokines/immunology , Fluorescent Antibody Technique , Macrophages/immunology , Macrophages/virology , Mice , Microscopy, Electron, Transmission , Phagosomes/immunology , Proteasome Endopeptidase Complex/immunology , Vacuoles/immunology
9.
Cell Microbiol ; 10(1): 100-11, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17651446

ABSTRACT

Inoculation of Leishmania (L.) spp. promastigotes in the dermis of mammals by blood-feeding sand flies can be accompanied by the rapid recruitment of neutrophils, inflammatory monocytes and dendritic cells. Despite the presence of these lytic leucocytes, parasitism is efficiently established. We show here that Leishmania donovani promastigotes are targeted to two different compartments in neutrophils. The compartments harbouring either damaged or non-damaged parasites were characterized at the electron microscopy (EM) level using the glucose 6-phosphatase cytochemistry and endosome-phagosome fusion assays. One involves the contribution of lysosomes leading to the formation of highly lytic compartments where parasites are rapidly degraded. The other is lysosome-independent and involves the contribution of a compartment displaying some features of the endoplasmic reticulum (ER) where parasites are protected from degradation. Using genetically modified parasites, we show that the promastigote surface lipophosphoglycan (LPG) is required to inhibit lysosome fusion and maintain parasites in neutrophil compartments displaying ER features. L. donovani-harbouring neutrophils that eventually enter apoptosis can be phagocytosed by macrophages enabling the stealth entry of parasites into their final replicative host cells. Thus, the ability of L. donovani to avoid trafficking into lysosomes-derived compartments in short-lived neutrophils constitutes a key process for the subsequent establishment of long-term parasitism.


Subject(s)
Endosomes/parasitology , Leishmania donovani/immunology , Lysosomes/parasitology , Macrophages/parasitology , Neutrophils/parasitology , Animals , Apoptosis/immunology , Cells, Cultured , Dogs , Endosomes/chemistry , Endosomes/ultrastructure , Glycosphingolipids/physiology , Lysosomes/chemistry , Lysosomes/ultrastructure , Mice , Microscopy, Electron, Transmission , Neutrophils/chemistry , Neutrophils/ultrastructure
10.
Cell ; 110(1): 119-31, 2002 Jul 12.
Article in English | MEDLINE | ID: mdl-12151002

ABSTRACT

Phagocytosis is a key aspect of our innate ability to fight infectious diseases. In this study, we have found that fusion of the endoplasmic reticulum (ER) with the macrophage plasmalemma, underneath phagocytic cups, is a source of membrane for phagosome formation in macrophages. Successive waves of ER become associated with maturing phagosomes during phagolysosome biogenesis. Thus, the ER appears to possess unexpectedly pluripotent fusion properties. ER-mediated phagocytosis is regulated in part by phosphatidylinositol 3-kinase and used for the internalization of inert particles and intracellular pathogens, regardless of their final trafficking in the host. In neutrophils, where pathogens are rapidly killed, the ER is not used as a major source of membrane for phagocytosis. We propose that intracellular pathogens have evolved to adapt and exploit ER-mediated phagocytosis to avoid destruction in host cells.


Subject(s)
Endoplasmic Reticulum/physiology , Macrophages/physiology , Phagocytosis/physiology , Phagosomes/physiology , Animals , Calcium-Binding Proteins/metabolism , Calnexin , Cell Membrane/physiology , Cells, Cultured , Dogs , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Intracellular Membranes/chemistry , Intracellular Membranes/physiology , Intracellular Membranes/ultrastructure , Macrophages/cytology , Phagosomes/metabolism , Phagosomes/ultrastructure , Phosphatidylinositol 3-Kinases/metabolism , Protein Transport/physiology , Proton-Translocating ATPases/metabolism , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL