Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Eur J Immunol ; 52(7): 1141-1157, 2022 07.
Article in English | MEDLINE | ID: mdl-35429346

ABSTRACT

IL-37 is an anti-inflammatory member of the IL-1 family that dampens inflammation associated with many noncommunicable diseases. However, mechanisms of IL-37 regulation remain understudied. We aimed to investigate the enzymatic cleavage of IL-37 that potentiates extracellular signalling, as well as pathways of IL-37 secretion. In human monocytes, mature IL-37 (mIL-37) was released following canonical NLRP3 inflammasome activation. The release of IL-37 was blocked by inhibiting plasma membrane permeability and in gasdermin-D-deficient THP-1 cells. While the cleavage of IL-37 was found to be constitutive, the release of mIL-37 was blocked in NLRP3-deficient THP-1 cells and by NLRP3 inhibitor MCC950 in THP-1s and primary human monocytes. IL-37 secretion also occurred after 18-h exposure to LPS, independently of the alternative NLRP3 inflammasome. This LPS-dependent IL-37 secretion required plasma membrane permeability, but not conventional protein secretion apparatus. Mutagenesis of the suggested caspase-1 cleavage site (D20) or the proposed alternative cleavage site (V46) did not completely block IL-37 processing. Therefore, we propose a novel pathway in which IL-37 is cleaved by caspase-1-independent mechanisms and released following canonical and alternative NLRP3 inflammasome triggers by differential pathways.


Subject(s)
Inflammasomes , Interleukin-1 , Monocytes , NLR Family, Pyrin Domain-Containing 3 Protein , Caspase 1/metabolism , Humans , Inflammasomes/metabolism , Interleukin-1/metabolism , Lipopolysaccharides , Monocytes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , THP-1 Cells
2.
Immunity ; 37(3): 487-500, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22981536

ABSTRACT

Cell volume regulation is a primitive response to alterations in environmental osmolarity. The NLRP3 inflammasome is a multiprotein complex that senses pathogen- and danger-associated signals. Here, we report that, from fish to mammals, the basic mechanisms of cell swelling and regulatory volume decrease (RVD) are sensed via the NLRP3 inflammasome. We found that a decrease in extracellular osmolarity induced a K(+)-dependent conformational change of the preassembled NLRP3-inactive inflammasome during cell swelling, followed by activation of the NLRP3 inflammasome and caspase-1, which was controlled by transient receptor potential channels during RVD. Both mechanisms were necessary for interleukin-1ß processing. Increased extracellular osmolarity prevented caspase-1 activation by different known NLRP3 activators. Collectively, our data identify cell volume regulation as a basic conserved homeostatic mechanism associated with the formation of the NLRP3 inflammasome and reveal a mechanism for NLRP3 inflammasome activation.


Subject(s)
Carrier Proteins/metabolism , Cell Size , Inflammasomes/metabolism , Macrophages/metabolism , Animals , Apoptosis Regulatory Proteins , Blotting, Western , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Caspase 1/genetics , Caspase 1/metabolism , Cell Line , Cells, Cultured , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , HEK293 Cells , Humans , Hypertonic Solutions/pharmacology , Interleukin-1beta/metabolism , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Macrophages/cytology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Osmolar Concentration , RNA Interference , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Time Factors
3.
Proc Natl Acad Sci U S A ; 115(40): E9371-E9380, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30232264

ABSTRACT

The NLRP3 inflammasome is an important regulator of inflammation and immunity. It is a multimolecular platform formed within cells that facilitates the activation of proinflammatory caspases to drive secretion of cytokines such as interleukin-1ß (IL-1ß). Knowledge of the mechanisms regulating formation of the NLRP3 inflammasome is incomplete. Here we report Cl- channel-dependent formation of dynamic ASC oligomers and inflammasome specks that remain inactive in the absence of K+ efflux. Formed after Cl- efflux exclusively, ASC specks are NLRP3 dependent, reversible, and inactive, although they further prime inflammatory responses, accelerating and enhancing release of IL-1ß in response to a K+ efflux-inducing stimulus. NEK7 is a specific K+ sensor and does not associate with NLRP3 under conditions stimulating exclusively Cl- efflux, but does after K+ efflux, activating the complex driving inflammation. Our investigation delivers mechanistic understanding into inflammasome activation and the regulation of inflammatory responses.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Chlorides/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Multimerization , Animals , CARD Signaling Adaptor Proteins/genetics , Female , Inflammasomes/genetics , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Ion Transport/genetics , Male , Mice , Mice, Knockout , NIMA-Related Kinases/genetics , NIMA-Related Kinases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Potassium/metabolism
4.
J Biol Chem ; 294(21): 8325-8335, 2019 05 24.
Article in English | MEDLINE | ID: mdl-30940725

ABSTRACT

Interleukin (IL)-1 family cytokines potently regulate inflammation, with the majority of the IL-1 family proteins being secreted from immune cells via unconventional pathways. In many cases, secretion of IL-1 cytokines appears to be closely coupled to cell death, yet the secretory mechanisms involved remain poorly understood. Here, we studied the secretion of the three best-characterized members of the IL-1 superfamily, IL-1α, IL-1ß, and IL-18, in a range of conditions and cell types, including murine bone marrow-derived and peritoneal macrophages, human monocyte-derived macrophages, HeLa cells, and mouse embryonic fibroblasts. We discovered that IL-1ß and IL-18 share a common secretory pathway that depends upon membrane permeability and can operate in the absence of complete cell lysis and cell death. We also found that the pathway regulating the trafficking of IL-1α is distinct from the pathway regulating IL-1ß and IL-18. Although the release of IL-1α could also be dissociated from cell death, it was independent of the effects of the membrane-stabilizing agent punicalagin, which inhibited both IL-1ß and IL-18 release. These results reveal that in addition to their role as danger signals released from dead cells, IL-1 family cytokines can be secreted in the absence of cell death. We propose that models used in the study of IL-1 release should be considered context-dependently.


Subject(s)
Bone Marrow Cells/metabolism , Interleukin-18/metabolism , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Macrophages, Peritoneal/metabolism , Animals , Bone Marrow Cells/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , HeLa Cells , Humans , Hydrolyzable Tannins/pharmacology , Macrophages, Peritoneal/cytology , Mice , Protein Transport/drug effects
5.
EMBO Rep ; 19(10)2018 10.
Article in English | MEDLINE | ID: mdl-30206189

ABSTRACT

The assembly and activation of the inflammasomes are tightly regulated by post-translational modifications, including ubiquitin. Deubiquitinases (DUBs) counteract the addition of ubiquitin and are essential regulators of immune signalling pathways, including those acting on the inflammasome. How DUBs control the assembly and activation of inflammasomes is unclear. Here, we show that the DUBs USP7 and USP47 regulate inflammasome activation in macrophages. Chemical inhibition of USP7 and USP47 blocks inflammasome formation, independently of transcription, by preventing ASC oligomerisation and speck formation. We also provide evidence that the ubiquitination status of NLRP3 itself is altered by inhibition of USP7 and USP47. Interestingly, we found that the activity of USP7 and USP47 increased in response to inflammasome activators. Using CRISPR/Cas9 in the macrophage cell line THP-1, we show that inflammasome activation is reduced when both USP7 and USP47 are knocked down. Altogether, these data reveal a new post-transcriptional role for USP47 and USP7 in inflammation by regulating inflammasome activation and the release of the pro-inflammatory cytokines IL-1ß and IL-18, and implicate dual USP7 and USP47 inhibitors as potential therapeutic agents for inflammatory disease.


Subject(s)
Inflammation/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Ubiquitin Thiolesterase/genetics , Ubiquitin-Specific Peptidase 7/genetics , CRISPR-Cas Systems/genetics , Deubiquitinating Enzymes/chemistry , Deubiquitinating Enzymes/genetics , Gene Knockdown Techniques , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , Inflammation/pathology , Interleukin-18/genetics , Interleukin-1beta/genetics , Macrophages/metabolism , Signal Transduction/genetics , Ubiquitin-Specific Proteases , Ubiquitination/genetics
6.
Int J Mol Sci ; 21(10)2020 May 15.
Article in English | MEDLINE | ID: mdl-32429301

ABSTRACT

Increasing evidence suggests that intestinal dysfunctions may represent early events in Alzheimer's disease and contribute to brain pathology. This study examined the relationship between onset of cognitive impairment and colonic dysfunctions in a spontaneous AD model before the full development of brain pathology. SAMP8 mice underwent Morris water maze and assessment of faecal output at four, six and eight months of age. In vitro colonic motility was examined. Faecal and colonic Aß, tau proteins, α-synuclein and IL-1ß were assessed by ELISA. Colonic citrate synthase activity was assessed by spectrophotometry. Colonic NLRP3, caspase-1 and ASC expression were evaluated by Western blotting. Colonic eosinophil density and claudin-1 expression were evaluated by immunohistochemistry. The effect of Aß on NLRP3 signalling and mitochondrial function was tested in cultured cells. Cognitive impairment and decreased faecal output occurred in SAMP8 mice from six months. When compared with SAMR1, SAMP8 animals displayed: (1) impaired in vitro colonic contractions; (2) increased enteric AD-related proteins, IL-1ß, active-caspase-1 expression and eosinophil density; and (3) decreased citrate synthase activity and claudin-1 expression. In THP-1 cells, Aß promoted IL-1ß release, which was abrogated upon incubation with caspase-1 inhibitor or in ASC-/- cells. Aß decreased mitochondrial function in THP-1 cells. In SAMP8, enteric AD-related proteins deposition, inflammation and impaired colonic excitatory neurotransmission, occurring before the full brain pathology development, could contribute to bowel dysmotility and represent prodromal events in AD.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Colon/pathology , Colon/physiopathology , Gastrointestinal Motility , Inflammation/pathology , Nerve Tissue Proteins/metabolism , Prodromal Symptoms , Amyloid beta-Peptides/metabolism , Animals , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/metabolism , Claudin-1/metabolism , Cognition , Eosinophils/pathology , Feces , Feeding Behavior , Humans , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Intestinal Mucosa/pathology , Mice , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Aggregates , THP-1 Cells , alpha-Synuclein/metabolism , tau Proteins/metabolism
7.
Immunology ; 155(3): 320-330, 2018 11.
Article in English | MEDLINE | ID: mdl-30098204

ABSTRACT

As a result of its strategic location, the epithelium is constantly exposed to a wide variety of pathogen and danger signals. Traditionally, the epithelium has been perceived as a defensive but passive barrier; however, it has now become evident that the epithelium senses and actively responds to these signals in order to maintain barrier homeostasis and contributes to the inflammatory response. One way it does this is by producing pro-inflammatory cytokines including interleukin-1ß (IL-1ß) and IL-18. These two cytokines are synthesized as inactive precursors, the maturation of which is mediated by pro-inflammatory caspases after the activation and assembly of macromolecular complexes called inflammasomes. Epithelial cells express a large panel of inflammasome components, and although the molecular mechanisms underlying the activation of these complexes in haematopoietic cells are well understood, how epithelial cells react to danger signals to activate the inflammasome remains unclear. We review and discuss how different inflammasomes contribute to barrier homeostasis and inflammation at several barrier sites, their mechanisms and how their aberrant regulation contributes to disease at the different epithelia.


Subject(s)
Epithelial Cells/immunology , Inflammasomes/immunology , Interleukin-18/immunology , Interleukin-1beta/immunology , Signal Transduction/immunology , Animals , Humans
8.
Immunol Cell Biol ; 95(2): 178-188, 2017 02.
Article in English | MEDLINE | ID: mdl-27559003

ABSTRACT

Infection and injury of the gut are associated with cell damage and release of molecules such as extracellular adenosine 5'-triphosphate (ATP), which is recognised by the purinergic P2X7 receptor (P2X7R). P2X7R is widely expressed in the gut by antigen-presenting cells (APCs) and epithelial cells, but the role of the P2X7R on epithelial cells is poorly understood. We investigated P2X7R in intestinal epithelium in vitro and in vivo using two model infections, Toxoplasma gondii and Trichinella spiralis. Lipopolysaccharide and ATP treatment of intestinal epithelial cells and infection with T. gondii in vitro did not promote inflammasome-associated interleukin-1ß (IL-1ß) or IL-18 secretion, but promoted C-C motif chemokine ligand 5 (CCL5), tumour necrosis factor-α and IL-6 production that were significantly reduced when the P2X7R was blocked. Similarly, in vivo, infection with either T. spiralis or T. gondii induced rapid upregulation of epithelial CCL5 in wild-type (wild-type (WT)) mice that was significantly reduced in P2X7R-/- littermate controls. The effects of reduced epithelial CCL5 were assayed by investigating recruitment of dendritic cells (DCs) to the epithelium. Infection induced a rapid recruitment of CD11c+CD103+ DC subsets into the epithelial layer of WT mice but not P2X7R-/- mice. In vitro chemotaxis assays and bone marrow chimeras demonstrated the importance of epithelial P2X7R in DC recruitment. P2X7R signalling in epithelial cells mediates chemokine responses to promote initiation of host immunity to infection.


Subject(s)
Gastrointestinal Tract/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Receptors, Purinergic P2X7/metabolism , Adaptive Immunity , Animals , Chemokine CCL5/biosynthesis , Chemotaxis , Dendritic Cells/metabolism , Epithelial Cells/metabolism , Epithelial Cells/parasitology , Intestinal Mucosa/pathology , Male , Mice, Inbred C57BL , Parasite Load , Receptors, Purinergic P2X7/deficiency , T-Lymphocytes/immunology , Toxoplasma , Toxoplasmosis/immunology , Toxoplasmosis/parasitology , Toxoplasmosis/pathology
10.
J Immunol ; 194(3): 1261-73, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25552542

ABSTRACT

Apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain (ASC) is a key adaptor molecule required for the inflammatory processes. ASC acts by bridging NLRP proteins, such as NLRP3, with procaspase-1 within the inflammasome complex, which subsequently results in the activation of caspase-1 and the secretion of IL-1ß and IL-18. In response to bacterial infection, ASC also forms specks by self-oligomerization to activate caspase-1 and induce pyroptosis. Hitherto, the role of these specks in NLRP3 inflammasome activation in response to danger signals, such as a hypotonic environment, largely has been unexplored. In this article, we report that, under hypotonic conditions and independently of NLRP3, ASC was able to form specks that did not activate caspase-1. These specks were not associated with pyroptosis and were controlled by transient receptor potential vanilloid 2 channel-mediated signaling. However, interaction with NLRP3 enhanced ASC speck formation, leading to fully functional inflammasomes and caspase-1 activation. This study reveals that the ASC speck can present different oligomerization assemblies and represents an essential step in the activation of functional NLRP3 inflammasomes.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Carrier Proteins/metabolism , Caspase 1/metabolism , Macrophages/metabolism , Animals , Apoptosis Regulatory Proteins/chemistry , CARD Signaling Adaptor Proteins , Calcium Channels/metabolism , Cell Line , Enzyme Activation , Humans , Inflammasomes/metabolism , Male , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Interaction Domains and Motifs , Protein Multimerization , Signal Transduction , TRPV Cation Channels/metabolism
11.
Mediators Inflamm ; 2016: 3481371, 2016.
Article in English | MEDLINE | ID: mdl-27597804

ABSTRACT

Inflammation is a protective response of the organism to tissue injury or infection. It occurs when the immune system recognizes Pathogen-Associated Molecular Patterns (PAMPs) or Damage-Associated Molecular Pattern (DAMPs) through the activation of Pattern Recognition Receptors. This initiates a variety of signalling events that conclude in the upregulation of proinflammatory molecules, which initiate an appropriate immune response. This response is tightly regulated since any aberrant activation of immune responses would have severe pathological consequences such as sepsis or chronic inflammatory and autoimmune diseases. Accumulative evidence shows that the ubiquitin system, and in particular ubiquitin-specific isopeptidases also known as deubiquitinases (DUBs), plays crucial roles in the control of these immune pathways. In this review we will give an up-to-date overview on the role of DUBs in the NF-κB pathway and inflammasome activation, two intrinsically related events triggered by activation of the membrane TLRs as well as the cytosolic NOD and NLR receptors. Modulation of DUB activity by small molecules has been proposed as a way to control dysregulation or overactivation of these key players of the inflammatory response. We will also discuss the advances and challenges of a potential use of DUBs as therapeutic targets in inflammatory pathologies.


Subject(s)
Deubiquitinating Enzymes/metabolism , Alarmins/metabolism , Animals , Humans , Inflammation/metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism , Signal Transduction/physiology
12.
J Biol Chem ; 289(51): 35582-92, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25371210

ABSTRACT

IL-1α and ß are key players in the innate immune system. The secretion of these cytokines by dendritic cells (DC) is integral to the development of proinflammatory responses. These cytokines are not secreted via the classical secretory pathway. Instead, 2 independent processes are required; an initial signal to induce up-regulation of the precursor pro-IL-1α and -ß, and a second signal to drive cleavage and consequent secretion. Pro-IL-1α and -ß are both cytosolic and thus, are potentially subject to post-translational modifications. These modifications may, in turn, have a functional outcome in the context of IL-1α and -ß secretion and hence inflammation. We report here that IL-1α and -ß were degraded intracellularly in murine bone marrow-derived DC and that this degradation was dependent on active cellular processes. In addition, we demonstrate that degradation was ablated when the proteasome was inhibited, whereas autophagy did not appear to play a major role. Furthermore, inhibition of the proteasome led to an accumulation of polyubiquitinated IL-1α and -ß, indicating that IL-1α and -ß were polyubiquitinated prior to proteasomal degradation. Finally, our investigations suggest that polyubiquitination and proteasomal degradation are not continuous processes but instead are up-regulated following DC activation. Overall, these data highlight that IL-1α and -ß polyubiquitination and proteasomal degradation are central mechanisms in the regulation of intracellular IL-1 levels in DC.


Subject(s)
Dendritic Cells/metabolism , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Polyubiquitin/metabolism , Proteasome Endopeptidase Complex/metabolism , Animals , Blotting, Western , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Dendritic Cells/drug effects , Female , Interleukin-1/genetics , Interleukin-1/metabolism , Interleukin-1alpha/genetics , Interleukin-1beta/genetics , Leupeptins/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred BALB C , Protein Precursors/genetics , Protein Precursors/metabolism , Proteolysis/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Ubiquitination/drug effects
13.
J Biol Chem ; 288(42): 30485-30494, 2013 Oct 18.
Article in English | MEDLINE | ID: mdl-24022484

ABSTRACT

The proinflammatory cytokine IL-1ß is a key mediator of inflammatory responses that contribute to and exacerbate brain injury. IL-1ß is synthesized by microglia in the brain as an inactive precursor (pro-IL-1ß). Cleavage of pro-IL-1ß to a mature form is stimulated by damage-associated molecular patterns (DAMPs). These DAMPs are sensed by a pattern recognition receptor called NLRP3, which forms an inflammasome, resulting in the activation of caspase-1 and cleavage of pro-IL-1ß. To date, regulation of the inflammasome in culture has been studied under normal culture conditions, and it is not known how DAMPs signal under disease relevant conditions such as acidosis. Given the presence of acidosis in pathological states, our objective was to test the hypothesis that acidic conditions modify DAMP-induced IL-1ß release from cultured primary mouse glial cells. When LPS-primed glial cells were stimulated with DAMPs under acidic conditions (pH 6.2), the predominant IL-1ß form secreted was the 20-kDa rather than the 17-kDa caspase-1-dependent species. Lactic acidosis, induced by the addition of 25 mm lactic acid, also induced the release of 20-kDa IL-1ß. This 20-kDa product was produced independently of NLRP3 and caspase-1 but was inhibited by the cathepsin D inhibitor pepstatin A. These data suggest that under disease relevant acidosis, DAMPs and lactic acid induce the secretion of IL-1ß independently of the inflammasome. Therapeutic strategies directed to the inhibition of IL-1ß processing should therefore consider alternative processing of IL-1ß in addition to caspase-1-dependent processing.


Subject(s)
Acidosis, Lactic/metabolism , Caspase 1/metabolism , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Neuroglia/metabolism , Signal Transduction , Acidosis, Lactic/chemically induced , Acidosis, Lactic/genetics , Acidosis, Lactic/pathology , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 1/genetics , Cathepsin D/antagonists & inhibitors , Cathepsin D/genetics , Cathepsin D/metabolism , Cell Line, Tumor , Humans , Hydrogen-Ion Concentration , Inflammasomes/genetics , Interleukin-1beta/genetics , Lactic Acid/adverse effects , Lactic Acid/pharmacology , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Neuroglia/pathology , Pepstatins/pharmacology , Protease Inhibitors/pharmacology
14.
J Biol Chem ; 288(4): 2721-33, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23209292

ABSTRACT

IL-1ß is a potent pro-inflammatory cytokine produced in response to infection or injury. It is synthesized as an inactive precursor that is activated by the protease caspase-1 within a cytosolic molecular complex called the inflammasome. Assembly of this complex is triggered by a range of structurally diverse damage or pathogen associated stimuli, and the signaling pathways through which these act are poorly understood. Ubiquitination is a post-translational modification essential for maintaining cellular homeostasis. It can be reversed by deubiquitinase enzymes (DUBs) that remove ubiquitin moieties from the protein thus modifying its fate. DUBs present specificity toward different ubiquitin chain topologies and are crucial for recycling ubiquitin molecules before protein degradation as well as regulating key cellular processes such as protein trafficking, gene transcription, and signaling. We report here that small molecule inhibitors of DUB activity inhibit inflammasome activation. Inhibition of DUBs blocked the processing and release of IL-1ß in both mouse and human macrophages. DUB activity was necessary for inflammasome association as DUB inhibition also impaired ASC oligomerization and caspase-1 activation without directly blocking caspase-1 activity. These data reveal the requirement for DUB activity in a key reaction of the innate immune response and highlight the therapeutic potential of DUB inhibitors for chronic auto-inflammatory diseases.


Subject(s)
Caspase 1/metabolism , Endopeptidases/physiology , Interleukin-1beta/metabolism , Animals , Carboxypeptidases/metabolism , Endopeptidases/chemistry , Fibroblast Growth Factor 1/metabolism , Fibroblast Growth Factor 2/metabolism , Humans , Hydrazones/pharmacology , Hydroxyurea/analogs & derivatives , Hydroxyurea/pharmacology , Immunity, Innate , Inflammation , Interleukin-1alpha/metabolism , Interleukins/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Protein Processing, Post-Translational , Protein Structure, Tertiary , Ubiquitin Thiolesterase
15.
Cell Death Discov ; 10(1): 354, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117604

ABSTRACT

NLRP3 forms a multiprotein inflammasome complex to initiate the inflammatory response when macrophages sense infection or tissue damage, which leads to caspase-1 activation, maturation and release of the inflammatory cytokines interleukin-1ß (IL-1ß) and IL-18 and Gasdermin-D (GSDMD) mediated pyroptosis. NLRP3 inflammasome activity must be controlled as unregulated and chronic inflammation underlies inflammatory and autoimmune diseases. Several findings uncovered that NLRP3 inflammasome activity is under the regulation of centrosome localized proteins such as NEK7 and HDAC6, however, whether the centrosome composition or structure is altered during the inflammasome activation is not known. Our data show that levels of the centrosomal scaffold protein pericentrin (PCNT) are reduced upon NLRP3 inflammasome activation via different activators in human and murine macrophages. PCNT loss occurs in the presence of membrane stabilizer punicalagin, suggesting this is not a consequence of membrane rupture. We found that PCNT loss is dependent on NLRP3 and active caspases as MCC950 and pan caspase inhibitor ZVAD prevent its degradation. Moreover, caspase-1 and GSDMD are both required for this NLRP3-mediated PCNT loss because absence of caspase-1 or GSDMD triggers an alternative regulation of PCNT via its cleavage by caspase-3 in response to nigericin stimulation. PCNT degradation occurs in response to nigericin, but also other NLRP3 activators including lysomotropic agent L-Leucyl-L-Leucine methyl ester (LLOMe) and hypotonicity but not AIM2 activation. Our work reveals that the NLRP3 inflammasome activation alters centrosome composition highlighting the need to further understand the role of this organelle during inflammatory responses.

16.
Life Sci Alliance ; 7(6)2024 Jun.
Article in English | MEDLINE | ID: mdl-38527803

ABSTRACT

Inflammasomes are immune complexes whose activation leads to the release of pro-inflammatory cytokines IL-18 and IL-1ß. Type I IFNs play a role in fighting infection and stimulate the expression of IFN-stimulated genes (ISGs) involved in inflammation. Despite the importance of these cytokines in inflammation, the regulation of inflammasomes by type I IFNs remains poorly understood. Here, we analysed RNA-sequencing data from patients with monogenic interferonopathies and found an up-regulation of several inflammasome-related genes. To investigate the effect of type I IFN on the inflammasome, we treated human monocyte-derived macrophages with IFN-α and observed an increase in CASP1 and GSDMD mRNA levels over time, whereas IL1B and NLRP3 were not directly correlated to IFN-α exposure time. IFN-α treatment reduced the release of mature IL-1ß and IL-18, but not caspase-1, in response to ATP-mediated NLRP3 inflammasome activation, suggesting regulation occurs at cytokine expression levels and not the inflammasome itself. However, more studies are required to investigate how regulation by IFN-α occurs and impacts NLRP3 and other inflammasomes at both transcriptional and post-translational levels.


Subject(s)
Interferon Type I , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Interferon Type I/metabolism , Interleukin-18/metabolism , Macrophages/metabolism , Cytokines/metabolism , Inflammation/metabolism , Caspase 1/metabolism
17.
Nat Commun ; 15(1): 6750, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117622

ABSTRACT

Interleukin-1α is a suggested dual-function cytokine that diverged from interleukin-1ß in mammals potentially by acquiring additional biological roles that relate to highly conserved regions in the pro-domain of interleukin-1α, including a nuclear localisation sequence and histone acetyltransferase-binding domains. Why evolution modified pro-interleukin-1α's subcellular location and protein interactome, and how this shaped interleukin-1α's intracellular role, is unknown. Here we show that TurboID proximity labelling with pro-interleukin-1α suggests a nuclear role for pro-interleukin-1α that involves interaction with histone acetyltransferases, including EP300. We also identify and validate inactivating mutations in the pro-interleukin-1α nuclear localisation sequence of multiple mammalian species, including toothed whales, castorimorpha and marsupials. However, histone acetyltransferase-binding domains are conserved in those species that have lost pro-interleukin-1α nuclear localisation. Together, these data suggest that histone acetyltransferase binding and nuclear localisation occurred together, and that while some species lost the nuclear localisation sequence in their pro-interleukin-1α, histone acetyltransferase binding ability was maintained. The nuclear localisation sequence was lost from several distinct species at different evolutionary times, suggesting convergent evolution, and that the loss of the nuclear localisation sequence confers some important biological outcome.


Subject(s)
Cell Nucleus , Evolution, Molecular , Interleukin-1alpha , Interleukin-1alpha/metabolism , Interleukin-1alpha/genetics , Animals , Cell Nucleus/metabolism , Humans , E1A-Associated p300 Protein/metabolism , E1A-Associated p300 Protein/genetics , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Protein Binding , Amino Acid Sequence
18.
Glia ; 61(5): 813-24, 2013 May.
Article in English | MEDLINE | ID: mdl-23404620

ABSTRACT

Macrophage can adopt several phenotypes, process call polarization, which is crucial for shaping inflammatory responses to injury. It is not known if microglia, a resident brain macrophage population, polarizes in a similar way, and whether specific microglial phenotypes modulate cell death in response to brain injury. In this study, we show that both BV2-microglia and mouse bone marrow derived macrophages (BMDMs) were able to adopt different phenotypes after LPS (M1) or IL-4 (M2) treatment in vitro, but regulated cell death differently when added to mouse organotypic hippocampal brain slices. BMDMs induced cell death when added to control slices and exacerbated damage when combined with oxygen-glucose deprivation (OGD), independently of their phenotype. In contrast, vehicle- and M2-BV2-microglia were protective against OGD-induced death. Direct treatment of brain slices with IL-4 (without cell addition) was protective against OGD and induced an M2 phenotype in the slice. In vivo, intracerebral injection of LPS or IL-4 in mice induced microglial phenotypes similar to the phenotypes observed in brain slices and in cultured cells. After injury induced by middle cerebral artery occlusion, microglial cells did not adopt classical M1/M2 phenotypes, suggesting that another subtype of regulatory phenotype was induced. This study highlights functional differences between macrophages and microglia, in response to brain injury with fundamentally different outcomes, even if both populations were able to adopt M1 or M2 phenotypes. These data suggest that macrophages infiltrating the brain from the periphery after an injury may be cytotoxic, independently of their phenotype, while microglia may be protective.


Subject(s)
Brain Injuries/metabolism , Brain Injuries/pathology , Glucose/deficiency , Hippocampus/pathology , Macrophages/metabolism , Macrophages/pathology , Microglia/metabolism , Microglia/pathology , Animals , Cell Death/physiology , Cell Hypoxia/physiology , Cells, Cultured , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Organ Specificity/physiology
19.
Eur J Immunol ; 42(3): 716-25, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22105559

ABSTRACT

Interleukin-1ß (IL-1ß) is a pro-inflammatory cytokine that regulates inflammatory responses to injury and infection. IL-1ß secretion requires the protease caspase-1, which is activated following recruitment to inflammasomes. Endogenous danger-associated molecular patterns (DAMPs) released from necrotic cells activate caspase-1 through an NLRP3-inflammasome. Here, we show that the endogenous lipid metabolite sphingosine (Sph) acts as a DAMP by inducing the NLRP3-inflammasome-dependent secretion of IL-1ß from macrophages. This process was dependent upon serine/threonine protein phosphatases since the PP1/PP2A inhibitors okadaic acid and calyculin A inhibited Sph-induced IL-1ß release. IL-1ß release induced by other well-characterized NLRP3-inflammasome activators, such as ATP and uric acid crystals, in addition to NLRC4 and AIM2 inflammasome activators was also blocked by these inhibitors. Thus, we propose Sph as a new DAMP, and that a serine/threonine phosphatase (PP1/PP2A)-dependent signal is central to the endogenous host mechanism through which diverse stimuli regulate inflammasome activation.


Subject(s)
Carrier Proteins/immunology , Inflammasomes/immunology , Interleukin-1beta/immunology , Peritonitis/immunology , Sphingosine/immunology , Animals , Blotting, Western , Enzyme Inhibitors/pharmacology , Flow Cytometry , Macrophages, Peritoneal/immunology , Marine Toxins , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Okadaic Acid/pharmacology , Oxazoles/pharmacology , Peritonitis/enzymology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/immunology , Random Allocation
20.
Nat Chem Biol ; 7(12): 902-8, 2011 Oct 23.
Article in English | MEDLINE | ID: mdl-22020553

ABSTRACT

The control of biochemical fluxes is distributed, and to perturb complex intracellular networks effectively it is often necessary to modulate several steps simultaneously. However, the number of possible permutations leads to a combinatorial explosion in the number of experiments that would have to be performed in a complete analysis. We used a multiobjective evolutionary algorithm to optimize reagent combinations from a dynamic chemical library of 33 compounds with established or predicted targets in the regulatory network controlling IL-1ß expression. The evolutionary algorithm converged on excellent solutions within 11 generations, during which we studied just 550 combinations out of the potential search space of ~9 billion. The top five reagents with the greatest contribution to combinatorial effects throughout the evolutionary algorithm were then optimized pairwise. A p38 MAPK inhibitor together with either an inhibitor of IκB kinase or a chelator of poorly liganded iron yielded synergistic inhibition of macrophage IL-1ß expression. Evolutionary searches provide a powerful and general approach to the discovery of new combinations of pharmacological agents with therapeutic indices potentially greater than those of single drugs.


Subject(s)
Algorithms , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Computer Simulation , Drug Discovery/methods , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cell Death/drug effects , Computational Biology/methods , Dose-Response Relationship, Drug , Humans , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/biosynthesis , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL