Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 11(1): 2331, 2020 05 11.
Article in English | MEDLINE | ID: mdl-32393780

ABSTRACT

Extracellular vesicles have an important function in cellular communication. Here, we show that human and mouse monocytes release TGF-ß1-transporting vesicles in response to the pathogenic fungus Candida albicans. Soluble ß-glucan from C. albicans binds to complement receptor 3 (CR3, also known as CD11b/CD18) on monocytes and induces the release of TGF-ß1-transporting vesicles. CR3-dependence is demonstrated using CR3-deficient (CD11b knockout) monocytes generated by CRISPR-CAS9 genome editing and isolated from CR3-deficient (CD11b knockout) mice. These vesicles reduce the pro-inflammatory response in human M1-macrophages as well as in whole blood. Binding of the vesicle-transported TGF-ß1 to the TGF-ß receptor inhibits IL1B transcription via the SMAD7 pathway in whole blood and induces TGFB1 transcription in endothelial cells, which is resolved upon TGF-ß1 inhibition. Notably, human complement-opsonized apoptotic bodies induce production of similar TGF-ß1-transporting vesicles in monocytes, suggesting that the early immune response might be suppressed through this CR3-dependent anti-inflammatory vesicle pathway.


Subject(s)
Immunomodulation , Macrophage-1 Antigen/metabolism , Monocytes/metabolism , Transforming Growth Factor beta1/metabolism , Transport Vesicles/metabolism , Animals , Apoptosis , Candida albicans/metabolism , Candida albicans/ultrastructure , Down-Regulation , Dynamic Light Scattering , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/pathology , Interleukin-6/genetics , Interleukin-6/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Models, Biological , Monocytes/microbiology , Monocytes/ultrastructure , Protein Transport , Solubility , Transcription, Genetic , Up-Regulation , beta-Glucans/metabolism
2.
J Innate Immun ; 10(2): 94-105, 2018.
Article in English | MEDLINE | ID: mdl-29237166

ABSTRACT

The human plasma contact system is an immune surveillance system activated by the negatively charged surfaces of bacteria and fungi and includes the kallikrein-kinin, the coagulation, and the fibrinolytic systems. Previous work shows that the contact system also activates complement, and that plasma enzymes like kallikrein, plasmin, thrombin, and FXII are involved in the activation process. Here, we show for the first time that kallikrein cleaves the central complement component C3 directly to yield active components C3b and C3a. The cleavage site within C3 is identical to that recognized by the C3 convertase. Also, kallikrein-generated C3b forms C3 convertases, which trigger the C3 amplification loop. Since kallikrein also cleaves factor B to yield Bb and Ba, kallikrein alone can trigger complement activation. Kallikrein-generated C3 convertases are inhibited by factor H; thus, the kallikrein activation pathway merges with the amplification loop of the alternative pathway. Taken together, these data suggest that activation of the contact system locally enhances complement activation on cell surfaces. The human pathogenic microbe Candida albicans activates the contact system in normal human serum. However, C. albicans immediately recruits factor H to the surface, thereby evading the alternative and likely kallikrein-mediated complement pathways.


Subject(s)
Complement Activation , Complement C3-C5 Convertases/metabolism , Complement C3/metabolism , Kallikreins/metabolism , Amino Acid Sequence , Animals , Candida albicans/immunology , Candidiasis/immunology , Candidiasis/microbiology , Cell Line, Transformed , Complement C3b/chemistry , Complement C3b/metabolism , Complement Factor B/metabolism , Complement Factor D/metabolism , Complement Factor H/pharmacology , Complement Pathway, Alternative , Factor XII/metabolism , Female , Humans , Immune Evasion , Mice, Inbred BALB C , Protein Binding/drug effects
3.
Front Immunol ; 8: 703, 2017.
Article in English | MEDLINE | ID: mdl-28663750

ABSTRACT

PURPOSE: The retinal pigment epithelium (RPE) is a main target for complement activation in age-related macular degeneration (AMD). The anaphylatoxins C3a and C5a have been thought to mostly play a role as chemoattractants for macrophages and immune cells; here, we explore whether they trigger RPE alterations. Specifically, we investigated the RPE as a potential immunoregulatory gate, allowing for active changes in the RPE microenvironment in response to complement. DESIGN: In vitro and in vivo analysis of signaling pathways. METHODS: Individual activities of and interaction between the two anaphylatoxin receptors were tested in cultured RPE cells by fluorescence microscopy, western blot, and immunohistochemistry. MAIN OUTCOME MEASURES: Intracellular free calcium, protein phosphorylation, immunostaining of tissues/cells, and multiplex secretion assay. RESULTS: Similar to immune cells, anaphylatoxin exposure resulted in increases in free cytosolic Ca2+, PI3-kinase/Akt activation, FoxP3 and FOXO1 phosphorylation, and cytokine/chemokine secretion. Differential responses were elicited depending on whether C3a and C5a were co-administered or applied consecutively, and response amplitudes in co-administration experiments ranged from additive to driven by C5a (C3a + C5a = C5a) or being smaller than those elicited by C3a alone (C3a + C5a < C3a). CONCLUSION: We suggest that this combination of integrative signaling between C3aR and C5aR helps the RPE to precisely adopt its immune regulatory function. These data further contribute to our understanding of AMD pathophysiology.

4.
Front Immunol ; 7: 671, 2016.
Article in English | MEDLINE | ID: mdl-28133459

ABSTRACT

Upon systemic infection with human pathogenic yeast Candida albicans (C. albicans), human monocytes and polymorph nuclear neutrophilic granulocytes are the first immune cells to respond and come into contact with C. albicans. Monocytes exert immediate candidacidal activity and inhibit germination, mediate phagocytosis, and kill fungal cells. Here, we show that human monocytes spontaneously respond to C. albicans cells via phagocytosis, decondensation of nuclear DNA, and release of this decondensed DNA in the form of extracellular traps (called monocytic extracellular traps: MoETs). Both subtypes of monocytes (CD14++CD16-/CD14+CD16+) formed MoETs within the first hours upon contact with C. albicans. MoETs were characterized by the presence of citrullinated histone, myeloperoxidase, lactoferrin, and elastase. MoETs were also formed in response to Staphylococcus aureus and Escherichia coli, indicating a general reaction of monocytes to infectious microbes. MoET induction differs from extracellular trap formation in macrophages as MoETs are not triggered by simvastatin, an inhibitor of cholesterol synthesis and inducer of extracellular traps in macrophages. Extracellular traps from both monocytes and neutrophils activate complement and C3b is deposited. However, factor H (FH) binds via C3b to the extracellular DNA, mediates cofactor activity, and inhibits the induction of the inflammatory cytokine interleukin-1 beta in monocytes. Altogether, the results show that human monocytes release extracellular DNA traps in response to C. albicans and that these traps finally bind FH via C3b to presumably support clearance without further inflammation.

SELECTION OF CITATIONS
SEARCH DETAIL
...