Elsevier

Current Opinion in Immunology

Volume 44, February 2017, Pages 7-13
Current Opinion in Immunology

Alternative inflammasome activation enables IL-1β release from living cells

https://doi.org/10.1016/j.coi.2016.10.007Get rights and content

Highlights

  • Alternative NLRP3 inflammasome activation comprises a distinct signaling entity.

  • Genetic and phenotypic criteria distinguish alternative and classical NLRP3 signaling.

  • This includes absence of pyroptosis and GSDMD-independent IL-1β secretion.

Classical modes of NLRP3 activation entail a priming step that enables its activation (signal 1) and a potassium efflux-dependent activation signal (signal 2) that triggers pyroptosome formation and pyroptosis, a lytic cell death necessary for IL-1β release. Opposing to that, human monocytes engage an alternative NLRP3 inflammasome pathway in response to LPS that proceeds in the absence of signal 2 activation and enables IL-1β secretion without pyroptosis. This specifically relies on Caspase-8 to propagate signaling to NLRP3, leading to inflammasome activation in absence of pyroptosome formation. Here, we summarize the current knowledge about alternative inflammasome activation, discuss potential extensions of this signaling entity beyond LPS-dependent activation, speculate about its role in tissue homeostasis and sterile inflammation and highlight the implications of pyroptosis-independent IL-1β secretion.

Introduction

The pleotropic cytokine IL-1β serves instrumental functions in initiation and orchestration of innate and adaptive immune responses. Its receptor IL-1R shares the intracellular TIR (Toll IL-1 receptor) signaling-domain with the well-characterized PRR-family of TLRs, thereby enabling TLR-like transcriptional responses in cells not expressing TLRs. This homology in signaling explains why the biological effects of IL-1β closely resemble activation of the innate immune system. IL-1β is a cytokine of numerous functions, exerting diverse effects on multiple cells types to culminate in broadly inflammatory events. Systemic consequences of IL-1β signaling include fever, vasodilatation, hypotension and acute phase response. Locally, IL-1 signaling enables mesenchymal and endothelial cells to express adhesion molecules facilitating recruitment of lymphocytes and myeloid cells. Consequently, the infiltrating immune cells are activated and triggered to further amplify inflammation. Besides its crucial roles in the induction and orchestration of innate immune responses, IL-1β has been implicated in adaptive responses by influencing Th1 and Th17-mediated immune activation. Aberrant IL-1β signaling has been associated with rare heritable monogenetic auto-inflammatory diseases like Cryopyrin-Associated Periodic Syndromes (CAPS) or Familial Mediterranean Fever (FMF), yet also with common multifactorial diseases, such as Type 2 diabetes or gout [1, 2, 3].

IL-1β belongs to the family of IL-1 cytokines, of which several members, including IL-1β and IL-18, are synthesized as inactive precursors within the cytosol, where they await proteolytic cleavage to gain bioactivity. This maturation step is most commonly executed by the cysteine protease Caspase-1, whose activity is controlled by a cytosolic activation platform called the inflammasome (Figure 1). At the same time, other enzymes, such as Caspase-8 or neutrophil proteases have been described to mature IL-1β. Inflammasome signaling entails a sensor molecule (e.g. NLRP1, NLRP3, NLRC4 or AIM2) that seeds the assembly of the adapter molecule ASC into helically structured filaments forming a high-molecular weight complex that is called the pyroptosome [4]. In turn, Caspase-1 becomes recruited to the pyroptosome to also form helical structures, which enable its proximity-induced proteolytical auto-activation. Once Caspase-1 has become matured into the active p102:p202 hetero-tetramer it is licensed to cleave the cytokine-precursors pro-IL-1β/pro-IL-18. Besides cytokine maturation, induction of a lytic type of cell death called pyroptosis constitutes a hallmark of inflammasome activation. This programmed cell death is mediated by Caspase-1-dependent cleavage of the pyroptosis-effector molecule GSDMD (Gasdermin-D). Caspase-1 maturation of GSDMD abolishes the intra-molecular auto-inhibition of the C-terminal domain [5••, 6••, 7••]. Subsequently, phospholipid binding specificities of the matured N-terminal fragment of GSDMD enable its translocation to the inner leaflet of the plasma membrane, where it forms round, pore-like structures of approximately 15 nm in diameter [8, 9, 10•, 11]. Consequently, breakdown of ion gradients over the plasma membrane leads to water influx driven by oncotic pressure, inducing the characteristic cell swelling and rupture of the plasma membrane. Although soluble cytosolic proteins are released during pyroptotic lysis, it has been shown that so called pore-induced intracellular traps (PITs) maintain larger fragments of the pyroptosed cell (including previously phagocytosed bacteria) within the cellular debris [12]. The previously noted correlation of pyroptosis and IL-1β release [13, 14] has been genetically supported by the defect in IL-1β secretion but not in maturation in murine macrophages deficient for GSDMD [5••, 6••, 7••]. To this end, the picture emerges that IL-1β secretion critically depends on GSDMD in the course of classical inflammasome pathways, either directly through the nascent pore or in the course of plasma membrane disintegration.

Section snippets

The NLRP3 inflammasome

NLRP3 is the most studied inflammasome sensor that has attracted much attention due to its role in anti-microbial defense, but also in sterile inflammatory conditions. Initially identified by its association with monogenetic, IL-1-mediated auto-inflammatory diseases (CAPS) [15], it has also been shown to play a central role in multi-factorial, widespread diseases like Type 2 diabetes, Alzheimer's disease, gout and atherosclerosis. A recently identified pharmacological inhibitor of NLRP3 [16]

LPS triggers alternative inflammasome activation in human monocytes in the absence of pyroptosis

In the course of inflammasome activation, the induction of pyroptosis and IL-1β secretion is strictly correlated and the requirement of lytic cell death for IL-1β secretion is well supported by genetic means [5••, 6••, 7••]. Apart from that, the proposed two-step model of NLRP3 activation has mainly been inferred from studies in murine macrophages that critically depend on signal 2 agonists for NLRP3 signaling. To this effect, not much attention has been paid to the fact that human monocytes

Broadening the concept of alternative inflammasome signaling

Notably, two additional studies have characterized NLRP3 activation patterns with striking similarities to alternative inflammasome activation, as observed in LPS-stimulated monocytes [42••, 43••] (Table 1). The first study investigated the role of the known immune-modulator oxPAPC, an endogenous oxidized membrane lipid, in inflammasome activation. In TLR-primed murine dendritic cells (DCs) oxPAPC is sufficient to activate a Caspase-11-NLRP3 inflammasome pathway that drives IL-1β secretion. For

Implications of alternative inflammasome activation

Ever since its discovery, the unconventional mode of secretion of IL-1β has been subject of intense research efforts. The notion that IL-1β is synthesized as an inactive precursor in the cytosol had already implied Golgi-independent secretion mechanisms. With the discovery of GSDMD it became clear that passive release either directly through the GSDMD pore or in the course of plasma membrane rupture constitutes the main mechanism of unconventional secretion of IL-1β during classical

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

This work was supported by the European Research Council (ERC-2014-CoG  647858  GENESIS) and the German Research Foundation (SFB 704).

References (59)

  • C.A. Dinarello

    A clinical perspective of IL-1b as the gatekeeper of inflammation

    Eur J Immunol

    (2011)
  • T. Fernandes-Alnemri et al.

    The pyroptosome: a supramolecular assembly of ASC dimers mediating inflammatory cell death via caspase-1 activation

    Cell Death Differ.

    (2007)
  • J. Shi et al.

    Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death

    Nature

    (2015)
  • N. Kayagaki et al.

    Caspase-11 cleaves gasdermin D for non-canonical inflammasome signaling

    Nature

    (2015)
  • W. He et al.

    Gasdermin D is an executor of pyroptosis and required for interleukin-1β secretion

    Cell Res

    (2015)
  • L. Sborgi et al.

    GSDMD pore formation in the plasma membrane constitutes the mechanism of pyroptotic cell death

    EMBO J

    (2016)
  • R.A. Aglietti et al.

    GsdmD p30 elicited by caspase-11 during pyroptosis forms pores in membranes

    Proc Natl Acad Sci

    (2016)
  • J. Ding et al.

    Pore-forming activity and structural autoinhibition of the gasdermin family

    Nature

    (2016)
  • X. Liu et al.

    Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores

    Nature

    (2016)
  • I. Jorgensen et al.

    Pyroptosis triggers pore-induced intracellular traps (PITs) that capture bacteria and lead to their clearance by efferocytosis [Internet]

    J Exp Med

    (2016)
  • F. Martín-sánchez et al.

    Inflammasome-dependent IL-1 β release depends upon membrane permeabilisation

    Cell Death Differ

    (2016)
  • T. Liu et al.

    Single-cell imaging of caspase-1 dynamics reveals an all-or-none inflammasome signaling response [Internet]

    Cell Rep

    (2013)
  • H.M. Hoffman et al.

    Mutation of a new gene encoding a putative pyrin-like protein causes familial cold autoinflammatory syndrome and Muckle-Wells syndrome. [Internet]

    Nat Genet

    (2001)
  • R.C. Coll et al.

    A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. [Internet]

    Nat Med

    (2015)
  • G.P.J. van Hout et al.

    The selective NLRP3-inflammasome inhibitor MCC950 reduces infarct size and preserves cardiac function in a pig model of myocardial infarction. [Internet]

    Eur Heart J

    (2016)
  • M.J. Primiano et al.

    Efficacy and pharmacology of the NLRP3 inflammasome inhibitor CP-456,773 (CRID3) in murine models of dermal and pulmonary inflammation [Internet]

    J Immunol

    (2016)
  • M.D. Tate et al.

    Reassessing the role of the NLRP3 inflammasome during pathogenic influenza A virus infection via temporal inhibition. [Internet]

    Sci Rep

    (2016)
  • V. Pétrilli et al.

    Activation of the NALP3 inflammasome is triggered by low intracellular potassium concentration

    Cell Death Differ

    (2007)
  • R. Muñoz-Planillo et al.

    K+ efflux is the common trigger of NLRP3 inflammasome activation by bacterial toxins and particulate matter. [Internet]

    Immunity

    (2013)
  • Cited by (92)

    • Gasdermin D kills bacteria

      2023, Microbiological Research
    • Prenatal-induced psychopathologies: All roads lead to microglia

      2023, Stress: Immunology and Inflammation: Handbook of Stress Series Volume 5
    • HIV-1 Gag and Vpr impair the inflammasome activation and contribute to the establishment of chronic infection in human primary macrophages

      2022, Molecular Immunology
      Citation Excerpt :

      However, this scenario may represent the dynamics of the initial events after HIV-1 infection in macrophages, contributing to the recruitment of other cell subtypes and the immunopathogenesis during viral infection. Another relevant aspect to be considered, as widely described elsewhere, the inflammasome also can be activated without inducing changes in the expression of NLRP3, AIM2 or RIG-I, but from direct proteolytic cleavage of Casp-1 that result in the release of IL-1α, IL-1β and IL-18 (Latz, 2010; Kugelberg, 2016; Gaidt and Hornung, 2017; Pyrillou et al., 2020). Recent studies (also using HIV-1Ba-L strain) explored the inflammasome activation in lineage or human primary macrophages.

    View all citing articles on Scopus
    View full text