S-acylation of NLRP3 provides a nigericin sensitive gating mechanism that controls access to the Golgi

Curation statements for this article:
  • Curated by eLife

    eLife logo

    eLife assessment

    This paper makes a valuable contribution by implicating S-acylation of Cys-130 in recruitment of the inflammasome receptor NLRP3 to the Golgi. Enzymes are identified as candidates for mediating S-acylation and de-acylation of NLRP3, and evidence is presented that S-acylation plays a role in response to the stress induced by nigericin treatment. Although it seems likely that Cys-130 does indeed contribute to membrane association of NLRP3, the mechanistic analyses are incomplete and the interpretations about the effects of nigericin are not fully conclusive.

This article has been Reviewed by the following groups

Read the full article See related articles

Abstract

NLRP3 is an inflammasome seeding pattern recognition receptor activated in response to multiple danger signals which perturb intracellular homeostasis. Electrostatic interactions between the NLRP3 polybasic (PB) region and negatively charged lipids on the trans-Golgi network (TGN) have been proposed to recruit NLRP3 to the TGN. In this study, we demonstrate that membrane association of NLRP3 is critically dependant on S-acylation of a highly conserved cysteine residue (Cys-130), which traps NLRP3 in a dynamic S-acylation cycle at the Golgi, and a series of hydrophobic residues preceding Cys-130 which act in conjunction with the PB region to facilitate Cys-130 dependent Golgi enrichment. Due to segregation from Golgi localised thioesterase enzymes caused by a nigericin induced breakdown in Golgi trafficking, NLRP3 becomes immobilised on the Golgi through reduced de-acylation of its Cys-130 lipid anchor, suggesting that disruptions in Golgi homeostasis are conveyed to NLRP3 through its acylation state. Thus, our work defines a nigericin sensitive S-acylation cycle that gates access of NLRP3 to the Golgi.

Article activity feed

  1. eLife assessment

    This paper makes a valuable contribution by implicating S-acylation of Cys-130 in recruitment of the inflammasome receptor NLRP3 to the Golgi. Enzymes are identified as candidates for mediating S-acylation and de-acylation of NLRP3, and evidence is presented that S-acylation plays a role in response to the stress induced by nigericin treatment. Although it seems likely that Cys-130 does indeed contribute to membrane association of NLRP3, the mechanistic analyses are incomplete and the interpretations about the effects of nigericin are not fully conclusive.

  2. Reviewer #1 (Public Review):

    This is an interesting study investigating the mechanisms underlying membrane targeting of the NLRP3 inflammasome and reporting a key role for the palmitoylation-depalmitoylation cycle of cys130 in NRLP3. The authors identify ZDHHC3 and APT2 as the specific ZDHHC and APT/ABHD enzymes that are responsible for the s-acylation and de-acylation of NLRP3, respectively. They show that the levels of ZDHHC3 and APT2, both localized at the Golgi, control the level of palmitoylation of NLRP3. The S-acylation-mediated membrane targeting of NLRP3 cooperates with polybasic domain (PBD)-mediated PI4P-binding to target NLRP3 to the TGN under steady-state conditions and to the disassembled TGN induced by the NLRP3 activator nigericin.

    However, the study has several weaknesses in its current form as outlined below.

    (1) The novelty of the findings concerning cys130 palmitoylation in NLRP3 is unfortunately compromised by recent reports on the acylation of different cysteines in NLRP3 (PMID: 38092000), including palmitoylation of the very same cys130 in NLRP3 (Yu et al https://doi.org/10.1101/2023.11.07.566005), which was shown to be relevant for NLRP3 activation in cell and animal models. What remains novel and intriguing is the finding that NLRP3 activators induce an imbalance in the acylation-deacylation cycle by segregating NLRP3 in late Golgi/endosomes from de-acylating enzymes confined in the Golgi. The interesting hypothesis put forward by the authors is that the increased palmitoylation of cys130 would finally contribute to the activation of NLRP3. However, the authors should clarify the trafficking pathway of acylated-NLRP3. This pathway should, in principle, coincide with that of TGN46 which constitutively recycles from the TGN to the plasma membrane and is trapped in endosomes upon treatment with nigericin.

    (2) To affect the S-acylation, the authors used 16 hrs treatment with 2-bromopalmitate (2-BP). In Figure 1f, it is quite clear that NLRP3 in 2-BP treated cells completely redistributed in spots dispersed throughout the cells upon nigericin treatment. What is the Golgi like in those cells? In other words, does 2-BP alter/affect Golgi morphology? What about PI4P levels after 2-BP treatment? These are important missing pieces of data since both the localization of many proteins and the activity of one key PI4K in the Golgi (i.e. PI4KIIalpha) are regulated by palmitoylation.

    (3) The authors argue that the spots observed with NLRP-GFP result from non-specific effects mediated by the addition of the GFP tag to the NLRP3 protein. However, puncta are visible upon nigericin treatment, as a hallmark of endosomal activation. How do the authors reconcile these data? Along the same lines, the NLRP3-C130S mutant behaves similarly to wt NLRP3 upon 2-BP treatment (Figure 1h). Are those NLRP3-C130S puncta positive for endosomal markers? Are they still positive for TGN46? Are they positive for PI4P?

    (4) The authors expressed the minimal NLRP3 region to identify the domain required for NLRP3 Golgi localization. These experiments were performed in control cells. It might be informative to perform the same experiments upon nigericin treatment to investigate the ability of NLRP3 to recognize activating signals. It has been reported that PI4P increases on Golgi and endosomes upon NG treatment. Hence, all the differences between the domains may be lost or preserved. In parallel, also the timing of such recruitment upon nigericin treatment (early or late event) may be informative for the dynamics of the process and of the contribution of the single protein domains.

    (5) As noted above for the chemical inhibitors (1) the authors should check the impact of altering the balance between acyl transferase and de-acylases on the Golgi organization and PI4P levels. What is the effect of overexpressing PATs on Golgi functions?

  3. Reviewer #2 (Public Review):

    Summary:

    This paper examines the recruitment of the inflammasome seeding pattern recognition receptor NLRP3 to the Golgi. Previously, electrostatic interactions between the polybasic region of NLRP3 and negatively charged lipids were implicated in membrane association. The current study reports that reversible S-acylation of the conserved Cys-130 residue, in conjunction with upstream hydrophobic residues plus the polybasic region, act together to promote Golgi localization of NLRP3, although additional parts of the protein are needed for full Golgi localization. Treatment with the bacterial ionophore nigericin inhibits membrane traffic and prevents Golgi-associated thioesterases from removing the acyl chain, causing NLRP3 to become immobilized at the Golgi. This mechanism is put forth as an explanation for how NLRP3 is activated in response to nigericin.

    Strengths:

    The experiments are generally well presented. It seems likely that Cys-130 does indeed play a previously unappreciated role in the membrane association of NLRP3.

    Weaknesses:

    The interpretations about the effects of nigericin are less convincing. Specific comments follow.

    (1) The experiments of Figure 4 bring into question whether Cys-130 is S-acylated. For Cys-130, S-acylation was seen only upon expression of a severely truncated piece of the protein in conjunction with overexpression of ZDHHC3. How do the authors reconcile this result with the rest of the story?

    (2) Nigericin seems to cause fragmentation and vesiculation of the Golgi. That effect complicates the interpretations. For example, the FRAP experiment of Figure 5 is problematic because the authors neglected to show that the FRAP recovery kinetics of non-acylated resident Golgi proteins are unaffected by nigericin. Similarly, the colocalization analysis in Figure 6 is less than persuasive when considering that nigericin significantly alters Golgi structure and could indirectly affect colocalization.