21 research outputs found

    Tumor necrosis factor is a necroptosis-associated alarmin

    Get PDF
    Necroptosis is a form of regulated cell death that can occur downstream of several immune pathways. While previous studies have shown that dysregulated necroptosis can lead to strong inflammatory responses, little is known about the identity of the endogenous molecules that trigger these responses. Using a reductionist in vitro model, we found that soluble TNF is strongly released in the context of necroptosis. On the one hand, necroptosis promotes TNF translation by inhibiting negative regulatory mechanisms acting at the post-transcriptional level. On the other hand, necroptosis markedly enhances TNF release by activating ADAM proteases. In studying TNF release at single-cell resolution, we found that TNF release triggered by necroptosis is activated in a switch-like manner that exceeds steady-state TNF processing in magnitude and speed. Although this shedding response precedes massive membrane damage, it is closely associated with lytic cell death. Further, we found that lytic cell death induction using a pore-forming toxin also triggers TNF shedding, indicating that the activation of ADAM proteases is not strictly related to the necroptotic pathway but likely associated with biophysical changes of the cell membrane upon lytic cell death. These results demonstrate that lytic cell death, particularly necroptosis, is a critical trigger for TNF release and thus qualify TNF as a necroptosis-associated alarmin

    Lack of PPARγ in Myeloid Cells Confers Resistance to Listeria monocytogenes Infection

    Get PDF
    The peroxisomal proliferator-activated receptor γ (PPARγ) is a nuclear receptor that controls inflammation and immunity. Innate immune defense against bacterial infection appears to be compromised by PPARγ. The relevance of PPARγ in myeloid cells, that organize anti-bacterial immunity, for the outcome of immune responses against intracellular bacteria such as Listeria monocytogenes in vivo is unknown. We found that Listeria monocytogenes infection of macrophages rapidly led to increased expression of PPARγ. This prompted us to investigate whether PPARγ in myeloid cells influences innate immunity against Listeria monocytogenes infection by using transgenic mice with myeloid-cell specific ablation of PPARγ (LysMCre×PPARγflox/flox). Loss of PPARγ in myeloid cells results in enhanced innate immune defense against Listeria monocytogenes infection both, in vitro and in vivo. This increased resistance against infection was characterized by augmented levels of bactericidal factors and inflammatory cytokines: ROS, NO, IFNγ TNF IL-6 and IL-12. Moreover, myeloid cell-specific loss of PPARγ enhanced chemokine and adhesion molecule expression leading to improved recruitment of inflammatory Ly6Chi monocytes to sites of infection. Importantly, increased resistance against Listeria infection in the absence of PPARγ was not accompanied by enhanced immunopathology. Our results elucidate a yet unknown regulatory network in myeloid cells that is governed by PPARγ and restrains both listeriocidal activity and recruitment of inflammatory monocytes during Listeria infection, which may contribute to bacterial immune escape. Pharmacological interference with PPARγ activity in myeloid cells might represent a novel strategy to overcome intracellular bacterial infection

    Self-guarding of MORC3 enables virulence factor-triggered immunity

    No full text
    Pathogens use virulence factors to inhibit the immune system1. The guard hypothesis2,3 postulates that hosts monitor (or 'guard') critical innate immune pathways such that their disruption by virulence factors provokes a secondary immune response1. Here we describe a 'self-guarded' immune pathway in human monocytes, in which guarding and guarded functions are combined in one protein. We find that this pathway is triggered by ICP0, a key virulence factor of herpes simplex virus type 1, resulting in robust induction of anti-viral type I interferon (IFN). Notably, induction of IFN by ICP0 is independent of canonical immune pathways and the IRF3 and IRF7 transcription factors. A CRISPR screen identified the ICP0 target MORC34 as an essential negative regulator of IFN. Loss of MORC3 recapitulates the IRF3- and IRF7-independent IFN response induced by ICP0. Mechanistically, ICP0 degrades MORC3, which leads to de-repression of a MORC3-regulated DNA element (MRE) adjacent to the IFNB1 locus. The MRE is required in cis for IFNB1 induction by the MORC3 pathway, but is not required for canonical IFN-inducing pathways. As well as repressing the MRE to regulate IFNB1, MORC3 is also a direct restriction factor of HSV-15. Our results thus suggest a model in which the primary anti-viral function of MORC3 is self-guarded by its secondary IFN-repressing function-thus, a virus that degrades MORC3 to avoid its primary anti-viral function will unleash the secondary anti-viral IFN response

    BAX/BAK-Induced Apoptosis Results in Caspase-8-Dependent IL-1β Maturation in Macrophages.

    Get PDF
    IL-1β is a cytokine of pivotal importance to the orchestration of inflammatory responses. Synthesized as an inactive pro-cytokine, IL-1β requires proteolytic maturation to gain biological activity. Here, we identify intrinsic apoptosis as a non-canonical trigger of IL-1β maturation. Guided by the discovery of the immunomodulatory activity of vioprolides, cyclic peptides isolated from myxobacteria, we observe IL-1β maturation independent of canonical inflammasome pathways, yet dependent on intrinsic apoptosis. Mechanistically, vioprolides inhibit MCL-1 and BCL2, which in turn triggers BAX/BAK-dependent mitochondrial outer membrane permeabilization (MOMP). Induction of MOMP results in the release of pro-apoptotic factors initiating intrinsic apoptosis, as well as the depletion of IAPs (inhibitors of apoptosis proteins). IAP depletion, in turn, operates upstream of ripoptosome complex formation, subsequently resulting in caspase-8-dependent IL-1β maturation. These results establish the ripoptosome/caspase-8 complex as a pro-inflammatory checkpoint that senses the perturbation of mitochondrial integrity

    Human Monocytes Engage an Alternative Inflammasome Pathway

    No full text
    Interleukin-1β (IL-1β) is a cytokine whose bioactivity is controlled by activation of the inflammasome. However, in response to lipopolysaccharide, human monocytes secrete IL-1β independently of classical inflammasome stimuli. Here, we report that this constituted a species-specific response that is not observed in the murine system. Indeed, in human monocytes, lipopolysaccharide triggered an “alternative inflammasome” that relied on NLRP3-ASC-caspase-1 signaling, yet was devoid of any classical inflammasome characteristics including pyroptosome formation, pyroptosis induction, and K+ efflux dependency. Genetic dissection of the underlying signaling pathway in a monocyte transdifferentiation system revealed that alternative inflammasome activation was propagated by TLR4-TRIF-RIPK1-FADD-CASP8 signaling upstream of NLRP3. Importantly, involvement of this signaling cascade was limited to alternative inflammasome activation and did not extend to classical NLRP3 activation. Because alternative inflammasome activation embraces both sensitivity and promiscuity of TLR4, we propose a pivotal role for this signaling cascade in TLR4-driven, IL-1β-mediated immune responses and immunopathology in humans.This work was supported by grants from the German Research Foundation (SFB704 and SFB670) to V.H. V.H. is a member of the Excellence cluster ImmunoSensation

    PPARγ expression is induced in bone-marrow derived monocytes after infection with <i>Listeria monocytogenes</i>.

    No full text
    <p>(A) Time course of PPARγ expression in human monocytes following <i>Listeria monocytogenes</i> infection detected by Western blot. (B) PPARγ target genes (n = 80) with the most significant differential expression in human macrophages post infection. Gene expression differences (log scale) are visualized as a heat map following hierarchical clustering of rows and columns (red = increased expression). (C,D) Time course of PPARγ expression in bone marrow derived macrophages (BMDM) post infection detected by Western blot or immunohistochemistry. (blue = DAPI staining the cell nucleus and <i>Listeria</i> DNA; green = PPARγ).</p

    Similar phagocytic capacity in myeloid cells from LysM- PPARγ<sup>WT</sup> and LysM- PPARγ<sup>KO</sup> mice.

    No full text
    <p>Bone marrow derived macrophages (BMDM) and peritoneal exudates cells (PEC) from LysM- PPARγ<sup>WT</sup> and LysM- PPARγ<sup>KO</sup> mice were infected with FITC-labelled <i>Listeria</i> at MOI 2 or MOI 5). (A) Mean fluorescence intensity (MFI) of FITC positive cells was assessed by flow cytometry. and (B) Percentage of <i>Listeria</i> containing macrophages (FITC positive). (C) BMDM or PEC from LysM- PPARγ<sup>WT</sup> and LysM- PPARγ<sup>KO</sup> mice were infected with <i>Listeria</i> (MOI 10) and intracellular <i>Listeria</i> growth was determined as CFU/ml in the lysates of infected cells. The <i>p</i> values for titres of WT vs. KO at 4 hrs p.i. were <0.05 and <0.01 for BMDM and PEC, respectively. (D–F) BMDM from LysM- PPARγ<sup>WT</sup> and LysM- PPARγ<sup>KO</sup> mice were infected with <i>Listeria</i> (MOI of 10). The capacity to produce reactive oxygen species (ROS) in <i>Listeria</i> infected macrophages was determined by the OxyBURST reagent (D). At 24 hrs p.i. the concentration of NO2- as surrogate marker for the production of reactive nitrogen intermediates was determined with Griess reagent (E). Expression of iNOS was analyzed by quantitative RT-PCR (F).</p
    corecore