169 results on '"M. Lamkanfi"'
Search Results
2. Author Correction: A human immune dysregulation syndrome characterized by severe hyperinflammation with a homozygous nonsense Roquin-1 mutation
- Author
-
S. J. Tavernier, V. Athanasopoulos, P. Verloo, G. Behrens, J. Staal, D. J. Bogaert, L. Naesens, M. De Bruyne, S. Van Gassen, E. Parthoens, J. Ellyard, J. Cappello, L. X. Morris, H. Van Gorp, G. Van Isterdael, Y. Saeys, M. Lamkanfi, P. Schelstraete, J. Dehoorne, V. Bordon, R. Van Coster, B. N. Lambrecht, B. Menten, R. Beyaert, C. G. Vinuesa, V. Heissmeyer, M. Dullaers, and F. Haerynck
- Subjects
Science - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2019
- Full Text
- View/download PDF
3. Author Correction: A human immune dysregulation syndrome characterized by severe hyperinflammation with a homozygous nonsense Roquin-1 mutation
- Author
-
Petra Schelstraete, Gesine Behrens, R. Van Coster, Bart N. Lambrecht, Eef Parthoens, G. Van Isterdael, H. Van Gorp, Rudi Beyaert, S. Van Gassen, Simon Tavernier, Melissa Dullaers, Filomeen Haerynck, Yvan Saeys, Jens Staal, M. Lamkanfi, Julia I. Ellyard, L. X. Morris, Patrick Verloo, Vigo Heissmeyer, Joke Dehoorne, Jean Cappello, Leslie Naesens, Victoria Bordon, Björn Menten, Carola G. Vinuesa, Delfien Bogaert, M. A. A. De Bruyne, and Vicki Athanasopoulos
- Subjects
0301 basic medicine ,Science ,media_common.quotation_subject ,Nonsense ,General Physics and Astronomy ,ComputerApplications_COMPUTERSINOTHERSYSTEMS ,02 engineering and technology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,ComputingMilieux_COMPUTERSANDEDUCATION ,Data_FILES ,medicine ,lcsh:Science ,media_common ,Genetics ,Multidisciplinary ,business.industry ,General Chemistry ,Immune dysregulation ,021001 nanoscience & nanotechnology ,030104 developmental biology ,Mutation (genetic algorithm) ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,lcsh:Q ,0210 nano-technology ,business - Abstract
textabstractThe original version of the Supplementary Information associated with this Article included an incorrect Supplementary Information file, in which only the first page of the file was included. The HTML has been updated to include a corrected and complete version of the Supplementary Information file.
- Published
- 2019
- Full Text
- View/download PDF
4. A human immune dysregulation syndrome characterized by severe hyperinflammation with a homozygous nonsense Roquin-1 mutation
- Author
-
Julia I. Ellyard, Carola G. Vinuesa, Jens Staal, L. X. Morris, Delfien Bogaert, H. Van Gorp, Björn Menten, Simon Tavernier, Jean Cappello, Victoria Bordon, Rudi Beyaert, Eef Parthoens, S. Van Gassen, Leslie Naesens, Bart N. Lambrecht, Gesine Behrens, M. Lamkanfi, Filomeen Haerynck, G. Van Isterdael, Vicki Athanasopoulos, R. Van Coster, Melissa Dullaers, Yvan Saeys, Patrick Verloo, Vigo Heissmeyer, Joke Dehoorne, Petra Schelstraete, M. A. A. De Bruyne, and Pulmonary Medicine
- Subjects
Male ,0301 basic medicine ,T-Lymphocytes ,medicine.medical_treatment ,General Physics and Astronomy ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,RNA decay ,HEMOPHAGOCYTIC LYMPHOHISTIOCYTOSIS ,Monocytes ,DISEASE ,Consanguinity ,Mice ,0302 clinical medicine ,DOMAIN ,Recurrence ,Medicine and Health Sciences ,lcsh:Science ,Receptor ,CONSTITUTIVE-DECAY ,Mutation ,Multidisciplinary ,MESSENGER-RNA DECAY ,Disease genetics ,Homozygote ,RNA-Binding Proteins ,Familial Hemophagocytic Lymphohistiocytosis ,3. Good health ,ROQ ,Cytokine ,Codon, Nonsense ,030220 oncology & carcinogenesis ,Cyclosporine ,Primary immunodeficiency disorders ,Tumor necrosis factor alpha ,Immunosuppressive Agents ,Adolescent ,Science ,Ubiquitin-Protein Ligases ,REGNASE-1 ,Article ,Lymphohistiocytosis, Hemophagocytic ,General Biochemistry, Genetics and Molecular Biology ,Immunophenotyping ,Inducible T-Cell Co-Stimulator Protein ,03 medical and health sciences ,Eosinophilia ,medicine ,Animals ,Humans ,Author Correction ,MACROPHAGE ACTIVATION SYNDROME ,Inflammation ,Hemophagocytic lymphohistiocytosis ,COMPLEX ,business.industry ,RECOGNITION ,Biology and Life Sciences ,General Chemistry ,biochemical phenomena, metabolism, and nutrition ,Receptors, OX40 ,Immune dysregulation ,medicine.disease ,030104 developmental biology ,ELEMENT ,HELPER T-CELLS ,Macrophage activation syndrome ,Immunology ,lcsh:Q ,business - Abstract
Hyperinflammatory syndromes are life-threatening disorders caused by overzealous immune cell activation and cytokine release, often resulting from defects in negative feedback mechanisms. In the quintessential hyperinflammatory syndrome familial hemophagocytic lymphohistiocytosis (HLH), inborn errors of cytotoxicity result in effector cell accumulation, immune dysregulation and, if untreated, tissue damage and death. Here, we describe a human case with a homozygous nonsense R688* RC3H1 mutation suffering from hyperinflammation, presenting as relapsing HLH. RC3H1 encodes Roquin-1, a posttranscriptional repressor of immune-regulatory proteins such as ICOS, OX40 and TNF. Comparing the R688* variant with the murine M199R variant reveals a phenotypic resemblance, both in immune cell activation, hypercytokinemia and disease development. Mechanistically, R688* Roquin-1 fails to localize to P-bodies and interact with the CCR4-NOT deadenylation complex, impeding mRNA decay and dysregulating cytokine production. The results from this unique case suggest that impaired Roquin-1 function provokes hyperinflammation by a failure to quench immune activation., Roquin-1 is a posttranscriptional regulator that controls the expression of many immune-related genes such as ICOS and TNFA. Here, the authors report a homozygous R688* loss of function mutation in Roquin-1 in a patient with syndromic uncontrolled hyperinflammation associated with immune cell activation and hypercytokinemia.
- Published
- 2019
5. Alice in caspase land. A phylogenetic analysis of caspases from worm to man
- Author
-
M Lamkanfi
- Subjects
Cell Biology ,Molecular Biology - Published
- 2002
- Full Text
- View/download PDF
6. SP0160 Protective and Detrimental Roles of Inflammasomes
- Author
-
M. Lamkanfi
- Subjects
Immunology ,Pyroptosis ,Familial Mediterranean fever ,Inflammasome ,Context (language use) ,Biology ,medicine.disease ,Pyrin domain ,General Biochemistry, Genetics and Molecular Biology ,Proinflammatory cytokine ,AIM2 ,Rheumatology ,NLRC4 ,medicine ,Immunology and Allergy ,medicine.drug - Abstract
Inflammasomes are multi-protein platforms that assemble in the cytosol of immune cells to cope with pathogens and cellular stress. The pattern-recognition receptors NLRP1, NLRP3, NLRC4, AIM2 and Pyrin all assemble canonical platforms for caspase-1 activation, and the related inflammatory caspases 4, 5 and 11 respond to cytosolic detection of the Gram-negative lipopolysaccharides. Inflammasomes contributes importantly to controlling pathogen replication through their roles in maturation and secretion of the inflammatory cytokines interleukin-(IL)1β and IL18, and they also induce pyroptosis. Contrasting to their beneficial roles in these host defense responses, deregulated inflammasome activation is thought to be detrimental in the context of chronic inflammatory arthritis and autoinflammatory diseases such as cryopyrin-associated periodic fever syndromes (CAPS) and Familial Mediterranean Fever (FMF). Disclosure of Interest None declared
- Published
- 2016
- Full Text
- View/download PDF
7. E11 Multiple inflammasomes direct innate immune responses against Salmonella
- Author
-
D.M. Monack, M. Lamkanfi, S. Mariathasan, P. Broz, K. Newton, and V.M. Dixit
- Subjects
Salmonella ,Innate immune system ,business.industry ,Innate lymphoid cell ,Immunology ,Medicine ,business ,medicine.disease_cause - Published
- 2010
- Full Text
- View/download PDF
8. A phylogenetic and functional overview of inflammatory caspases and caspase-1-related CARD-only proteins.
- Author
-
K. Kersse, T. Vanden Berghe, M. Lamkanfi, and P. Vandenabeele
- Subjects
CYSTEINE proteinases ,MOLECULAR phylogeny ,INFLAMMATION ,CYTOKINES ,NF-kappa B ,LITERATURE reviews ,HUMAN genome - Abstract
Caspase 1 is a cysteinyl aspartate-specific proteinase involved in the maturation of inflammatory cytokines such as pro-IL-1β (interleukin-1β) and pro-IL-18. Caspase 1 clusters phylogenetically together with human caspases 4, 5 and 12 and murine caspases 11 and 12, and forms the group of the so-called inflammatory caspases. Caspase 1 consists of an N-terminal CARD (caspase recruitment domain) and a proteolytic domain containing the catalytic residues. The CARD-containing prodomain is involved in the formation of the protease-activating inflammasome complex. We have also found that the prodomain is necessary and sufficient for the activation of NF-κB (nuclear factor κB). The human genome also contains three caspase-1-related CARD-only decoy proteins [COP (CARD-only protein), INCA (inhibitory CARD) and ICEBERG], which are located near the caspase 1 locus. In this mini-review, we focus on the evolutionary aspects of the inflammatory caspase locus in the human, chimpanzee, Rhesus monkey, mouse and rat. Furthermore, we discuss the functional characteristics of the caspase-1-related CARD-only proteins in relation to caspase-1-mediated IL-1β maturation and NF-κB activation. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
9. Infection history imprints prolonged changes to the epigenome, transcriptome and function of Kupffer cells.
- Author
-
Musrati MA, Stijlemans B, Azouz A, Kancheva D, Mesbahi S, Hadadi E, Lebegge E, Ali L, De Vlaminck K, Scheyltjens I, Vandamme N, Zivalj M, Assaf N, Elkrim Y, Ahmidi I, Huart C, Lamkanfi M, Guilliams M, De Baetselier P, Goriely S, Movahedi K, and Van Ginderachter JA
- Subjects
- Animals, Mice, Trypanosoma brucei brucei genetics, Trypanosomiasis parasitology, Macrophages metabolism, Macrophages parasitology, Disease Models, Animal, Kupffer Cells metabolism, Transcriptome, Epigenome, Liver parasitology, Liver metabolism
- Abstract
Background & Aims: Liver macrophages fulfill various homeostatic functions and represent an essential line of defense against pathogenic insults. However, it remains unclear whether a history of infectious disease in the liver leads to long-term alterations to the liver macrophage compartment., Methods: We utilized a curable model of parasitic infection invoked by the protozoan parasite Trypanosoma brucei brucei to investigate whether infection history can durably reshape hepatic macrophage identity and function. Employing a combination of fate mapping, single-cell CITE-sequencing, single-nuclei multiome analysis, epigenomic analysis, and functional assays, we studied the alterations to the liver macrophage compartment during and after the resolution of infection., Results: We show that T. brucei brucei infection alters the composition of liver-resident macrophages, leading to the infiltration of monocytes that differentiate into various infection-associated macrophage populations with divergent transcriptomic profiles. Whereas infection-associated macrophages disappear post-resolution of infection, monocyte-derived macrophages engraft in the liver, assume a Kupffer cell (KC)-like profile and co-exist with embryonic KCs in the long-term. Remarkably, the prior exposure to infection imprinted an altered transcriptional program on post-resolution KCs that was underpinned by an epigenetic remodeling of KC chromatin landscapes and a shift in KC ontogeny, along with transcriptional and epigenetic alterations in their niche cells. This reprogramming altered KC functions and was associated with increased resilience to a subsequent bacterial infection., Conclusion: Our study demonstrates that a prior exposure to a parasitic infection induces trained immunity in KCs, reshaping their identity and function in the long-term., Impact and Implications: Although the liver is frequently affected during infections, and despite housing a major population of resident macrophages known as Kupffer cells (KCs), it is currently unclear whether infections can durably alter KCs and their niche cells. Our study provides a comprehensive investigation into the long-term impact of a prior, cured parasitic infection, unveiling long-lasting ontogenic, epigenetic, transcriptomic and functional changes to KCs as well as KC niche cells, which may contribute to KC remodeling. Our data suggest that infection history may continuously reprogram KCs throughout life with potential implications for subsequent disease susceptibility in the liver, influencing preventive and therapeutic approaches., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
10. Author Correction: Drugging the NLRP3 inflammasome: from signalling mechanisms to therapeutic targets.
- Author
-
Vande Walle L and Lamkanfi M
- Published
- 2024
- Full Text
- View/download PDF
11. NLRP3 inflammasome activation and pyroptosis are dispensable for tau pathology.
- Author
-
Paesmans I, Van Kolen K, Vandermeeren M, Shih PY, Wuyts D, Boone F, Garcia Sanchez S, Grauwen K, Van Hauwermeiren F, Van Opdenbosch N, Lamkanfi M, van Loo G, and Bottelbergs A
- Abstract
Background: Neuroinflammation is widely recognized as a key factor in the pathogenesis of Alzheimer's disease (AD), alongside ß-amyloid deposition and the formation of neurofibrillary tangles. The NLR family pyrin domain containing 3 (NLRP3) inflammasome, part of the innate immune system, has been implicated in the neuropathology of both preclinical amyloid and tau transgenic models. Activation of the NLRP3 pathway involves an initial priming step, which increases the expression of Nlrp3 and interleukin (IL)-1β, followed by the assembly of the NLRP3 inflammasome complex, comprising NLRP3, ASC, and caspase-1. This assembly leads to the proteolytic maturation of the pro-inflammatory cytokines IL-1β and IL-18. Additionally, the NLRP3 inflammasome induces Gasdermin D (GSDMD) cleavage, forming membrane pores through which IL-1β and IL-18 are secreted. Inhibition of NLRP3 has been shown to enhance plaque clearance by modulating microglial activation. Furthermore, blocking NLRP3 in tau transgenic mice has been found to reduce tau phosphorylation by affecting the activity of certain tau kinases and phosphatases., Methods: In this study, organotypic brain slice cultures from P301S transgenic mice were treated with lipopolysaccharide (LPS) plus nigericin as a positive control or exposed to tau seeds (K18) to evaluate NLRP3 inflammasome activation. The effect of tau seeding on NLRP3 activity was further examined using Meso Scale Discovery (MSD) assays to measure IL1β secretion levels in the presence and absence of NLRP3 inhibitors. The role of NLRP3 activity was investigated in full-body Nlrp3 knockout mice crossbred with the tau transgenic P301S model. Additionally, full-body and microglia-selective Gsdmd knockout mice were crossbred with P301S mice, and tau pathology and neurodegeneration were evaluated at early and late stages of the disease using immunohistochemistry and biochemical assays., Results: Activation of the NLRP3 pathway was observed in the mouse organotypic slice culture (OSC) model following stimulation with LPS and nigericin or exposure to tau seeds. However, Nlrp3 deficiency did not mitigate tauopathy or neurodegeneration in P301S mice in vivo , showing only a minor effect on plasma neurofilament (NF-L) levels. Consistently, Gsdmd deficiency did not alter tau pathology in P301S mice. Furthermore, neither full-body nor microglia-selective Gsdmd deletion had an impact on neuronal pathology or the release of pro-inflammatory cytokines., Conclusion: The absence of key components of the NLRP3 inflammasome pathway did not yield a beneficial effect on tau pathology or neurodegeneration in the preclinical Tau-P301S mouse model of AD. Nonetheless, organotypic slice cultures could serve as a valuable ex vivo mechanistic model for evaluating NLRP3 pathway activation and pharmacological inhibitors., Competing Interests: AB, DW, IP, KG, KV, and NO are employees of Jansssssen Pharmaceutica. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Paesmans, Van Kolen, Vandermeeren, Shih, Wuyts, Boone, Garcia Sanchez, Grauwen, Van Hauwermeiren, Van Opdenbosch, Lamkanfi, van Loo and Bottelbergs.)
- Published
- 2024
- Full Text
- View/download PDF
12. Oxylipins and metabolites from pyroptotic cells act as promoters of tissue repair.
- Author
-
Mehrotra P, Maschalidi S, Boeckaerts L, Maueröder C, Tixeira R, Pinney J, Burgoa Cardás J, Sukhov V, Incik Y, Anderson CJ, Hu B, Keçeli BN, Goncalves A, Vande Walle L, Van Opdenbosch N, Sergushichev A, Hoste E, Jain U, Lamkanfi M, and Ravichandran KS
- Subjects
- Animals, Female, Humans, Mice, Caspase 1 metabolism, Cell Movement, Cell Proliferation, Cyclooxygenase 2 metabolism, Dinoprostone biosynthesis, Dinoprostone metabolism, Fibroblasts metabolism, Fibroblasts cytology, Gasdermins metabolism, Inflammasomes metabolism, Interleukin-1beta, Lipidomics, Mice, Inbred C57BL, Phosphate-Binding Proteins metabolism, Macrophages metabolism, Macrophages cytology, Oxylipins metabolism, Pyroptosis, Secretome metabolism, Wound Healing physiology
- Abstract
Pyroptosis is a lytic cell death mode that helps limit the spread of infections and is also linked to pathology in sterile inflammatory diseases and autoimmune diseases
1-4 . During pyroptosis, inflammasome activation and the engagement of caspase-1 lead to cell death, along with the maturation and secretion of the inflammatory cytokine interleukin-1β (IL-1β). The dominant effect of IL-1β in promoting tissue inflammation has clouded the potential influence of other factors released from pyroptotic cells. Here, using a system in which macrophages are induced to undergo pyroptosis without IL-1β or IL-1α release (denoted Pyro-1 ), we identify unexpected beneficial effects of the Pyro-1 secretome. First, we noted that the Pyro-1 supernatants upregulated gene signatures linked to migration, cellular proliferation and wound healing. Consistent with this gene signature, Pyro-1 supernatants boosted migration of primary fibroblasts and macrophages, and promoted faster wound closure in vitro and improved tissue repair in vivo. In mechanistic studies, lipidomics and metabolomics of the Pyro-1 supernatants identified the presence of both oxylipins and metabolites, linking them to pro-wound-healing effects. Focusing specifically on the oxylipin prostaglandin E2 (PGE2 ), we find that its synthesis is induced de novo during pyroptosis, downstream of caspase-1 activation and cyclooxygenase-2 activity; further, PGE2 synthesis occurs late in pyroptosis, with its release dependent on gasdermin D pores opened during pyroptosis. As for the pyroptotic metabolites, they link to immune cell infiltration into the wounds, and polarization to CD301+ macrophages. Collectively, these data advance the concept that the pyroptotic secretome possesses oxylipins and metabolites with tissue repair properties that may be harnessed therapeutically., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
13. Dual neutrophil subsets exacerbate or suppress inflammation in tuberculosis via IL-1β or PD-L1.
- Author
-
Doz-Deblauwe E, Bounab B, Carreras F, Fahel JS, Oliveira SC, Lamkanfi M, Le Vern Y, Germon P, Pichon J, Kempf F, Paget C, Remot A, and Winter N
- Subjects
- Animals, Mice, Mice, Inbred C57BL, Mice, Knockout, Mycobacterium tuberculosis immunology, Disease Models, Animal, Female, Humans, B7-H1 Antigen metabolism, B7-H1 Antigen genetics, Neutrophils immunology, Neutrophils metabolism, Interleukin-1beta metabolism, Inflammation immunology, Inflammation metabolism, Tuberculosis immunology, Tuberculosis microbiology, Tuberculosis metabolism, Lung immunology, Lung microbiology, Lung metabolism, Lung pathology
- Abstract
Neutrophils can be beneficial or deleterious during tuberculosis (TB). Based on the expression of MHC-II and programmed death ligand 1 (PD-L1), we distinguished two functionally and transcriptionally distinct neutrophil subsets in the lungs of mice infected with mycobacteria. Inflammatory [MHC-II
- , PD-L1lo ] neutrophils produced inflammasome-dependent IL-1β in the lungs in response to virulent mycobacteria and "accelerated" deleterious inflammation, which was highly exacerbated in IFN-γR-/- mice. Regulatory [MHC-II+ , PD-L1hi ] neutrophils "brake" inflammation by suppressing T-cell proliferation and IFN-γ production. Such beneficial regulation, which depends on PD-L1, is controlled by IFN-γR signaling in neutrophils. The hypervirulent HN878 strain from the Beijing genotype curbed PD-L1 expression by regulatory neutrophils, abolishing the braking function and driving deleterious hyperinflammation in the lungs. These findings add a layer of complexity to the roles played by neutrophils in TB and may explain the reactivation of this disease observed in cancer patients treated with anti-PD-L1., (© 2024 Doz-Deblauwe et al.)- Published
- 2024
- Full Text
- View/download PDF
14. Nlrp2 deletion ameliorates kidney damage in a mouse model of cystinosis.
- Author
-
Rossi MN, Matteo V, Diomedi-Camassei F, De Leo E, Devuyst O, Lamkanfi M, Caiello I, Loricchio E, Bellomo F, Taranta A, Emma F, De Benedetti F, and Prencipe G
- Subjects
- Animals, Cystine metabolism, Kidney pathology, RNA, Messenger, Disease Models, Animal, Mice, Cystinosis genetics, Cystinosis metabolism, Cystinosis pathology, Kidney Diseases pathology, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Apoptosis Regulatory Proteins genetics, Apoptosis Regulatory Proteins metabolism
- Abstract
Cystinosis is a rare autosomal recessive disorder caused by mutations in the CTNS gene that encodes cystinosin, a ubiquitous lysosomal cystine/H
+ antiporter. The hallmark of the disease is progressive accumulation of cystine and cystine crystals in virtually all tissues. At the kidney level, human cystinosis is characterized by the development of renal Fanconi syndrome and progressive glomerular and interstitial damage leading to end-stage kidney disease in the second or third decade of life. The exact molecular mechanisms involved in the pathogenesis of renal disease in cystinosis are incompletely elucidated. We have previously shown upregulation of NLRP2 in human cystinotic proximal tubular epithelial cells and its role in promoting inflammatory and profibrotic responses. Herein, we have investigated the role of NLRP2 in vivo using a mouse model of cystinosis in which we have confirmed upregulation of Nlrp2 in the renal parenchyma. Our studies show that double knock out Ctns-/- Nlrp2-/- animals exhibit delayed development of Fanconi syndrome and kidney tissue damage. Specifically, we observed at 4-6 months of age that animals had less glucosuria and calciuria and markedly preserved renal tissue, as assessed by significantly lower levels of inflammatory cell infiltration, tubular atrophy, and interstitial fibrosis. Also, the mRNA expression of some inflammatory mediators ( Cxcl1 and Saa1 ) and the rate of apoptosis were significantly decreased in 4-6-month old kidneys harvested from Ctns-/- Nlrp2-/- mice compared to those obtained from Ctns-/- mice. At 12-14 months of age, renal histological was markedly altered in both genetic models, although double KO animals had lower degree of polyuria and low molecular weight proteinuria and decreased mRNA expression levels of Il6 and Mcp1 . Altogether, these data indicate that Nlrp2 is a potential pharmacological target for delaying progression of kidney disease in cystinosis., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Rossi, Matteo, Diomedi-Camassei, De Leo, Devuyst, Lamkanfi, Caiello, Loricchio, Bellomo, Taranta, Emma, De Benedetti and Prencipe.)- Published
- 2024
- Full Text
- View/download PDF
15. Novel chemotype NLRP3 inhibitors that target the CRID3-binding pocket with high potency.
- Author
-
Vande Walle L, Said M, Paerewijck O, Bertoni A, Gattorno M, Linclau B, and Lamkanfi M
- Subjects
- Humans, Animals, Mice, Sulfonamides pharmacology, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Inflammasomes metabolism
- Abstract
The NLRP3 inflammasome plays a central role in various human diseases. Despite significant interest, most clinical-grade NLRP3 inhibitors are derived from sulfonylurea inhibitor CRID3 (also called MCC950). Here, we describe a novel chemical class of NLRP3-inhibiting compounds (NIC) that exhibit potent and selective NLRP3 inflammasome inhibition in human monocytes and mouse macrophages. BRET assays demonstrate that they physically interact with NLRP3. Structural modeling further reveals they occupy the same binding site of CRID3 but in a critically different conformation. Furthermore, we show that NIC-11 and NIC-12 lack the off-target activity of CRID3 against the enzymatic activity of carbonic anhydrases I and II. NIC-12 selectively reduces circulating IL-1ß levels in the LPS-endotoxemia model in mice and inhibits NLRP3 inflammasome activation in CAPS patient monocytes and mouse macrophages with about tenfold increased potency compared with CRID3. Altogether, this study unveils a new chemical class of highly potent and selective NLRP3-targeted inhibitors with a well-defined molecular mechanism to complement existing CRID3-based NLRP3 inhibitors in pharmacological studies and serve as novel chemical leads for the development of NLRP3-targeted therapies., (© 2024 Vande Walle et al.)
- Published
- 2024
- Full Text
- View/download PDF
16. Inflammasome signaling is dispensable for ß-amyloid-induced neuropathology in preclinical models of Alzheimer's disease.
- Author
-
Srinivasan S, Kancheva D, De Ren S, Saito T, Jans M, Boone F, Vandendriessche C, Paesmans I, Maurin H, Vandenbroucke RE, Hoste E, Voet S, Scheyltjens I, Pavie B, Lippens S, Schwabenland M, Prinz M, Saido T, Bottelbergs A, Movahedi K, Lamkanfi M, and van Loo G
- Subjects
- Mice, Animals, Inflammasomes, NLR Family, Pyrin Domain-Containing 3 Protein genetics, Amyloid beta-Peptides, Amyloid beta-Protein Precursor genetics, Neuroinflammatory Diseases, Mice, Transgenic, Amyloid, Amyloidogenic Proteins, Alzheimer Disease pathology
- Abstract
Background: Alzheimer's disease (AD) is the most common neurodegenerative disorder affecting memory and cognition. The disease is accompanied by an abnormal deposition of ß-amyloid plaques in the brain that contributes to neurodegeneration and is known to induce glial inflammation. Studies in the APP/PS1 mouse model of ß-amyloid-induced neuropathology have suggested a role for inflammasome activation in ß-amyloid-induced neuroinflammation and neuropathology., Methods: Here, we evaluated the in vivo role of microglia-selective and full body inflammasome signalling in several mouse models of ß-amyloid-induced AD neuropathology., Results: Microglia-specific deletion of the inflammasome regulator A20 and inflammasome effector protease caspase-1 in the App
NL-G-F and APP/PS1 models failed to identify a prominent role for microglial inflammasome signalling in ß-amyloid-induced neuropathology. Moreover, global inflammasome inactivation through respectively full body deletion of caspases 1 and 11 in AppNL-G-F mice and Nlrp3 deletion in APP/PS1 mice also failed to modulate amyloid pathology and disease progression. In agreement, single-cell RNA sequencing did not reveal an important role for Nlrp3 signalling in driving microglial activation and the transition into disease-associated states, both during homeostasis and upon amyloid pathology., Conclusion: Collectively, these results question a generalizable role for inflammasome activation in preclinical amyloid-only models of neuroinflammation., Competing Interests: SD, IP, HM, and AB are employed by Janssen Pharmaceutica NV. ML serves as a consultant for Ventyx Biosciences and Novo Nordisk outside of the submitted work. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Srinivasan, Kancheva, De Ren, Saito, Jans, Boone, Vandendriessche, Paesmans, Maurin, Vandenbroucke, Hoste, Voet, Scheyltjens, Pavie, Lippens, Schwabenland, Prinz, Saido, Bottelbergs, Movahedi, Lamkanfi and van Loo.)- Published
- 2024
- Full Text
- View/download PDF
17. Drugging the NLRP3 inflammasome: from signalling mechanisms to therapeutic targets.
- Author
-
Vande Walle L and Lamkanfi M
- Subjects
- Humans, Signal Transduction, Inflammation drug therapy, Inflammation metabolism, Cytokines metabolism, Nerve Growth Factors, Cell Adhesion Molecules, Neuronal, Inflammasomes metabolism, NLR Family, Pyrin Domain-Containing 3 Protein metabolism
- Abstract
Diseases associated with chronic inflammation constitute a major health burden across the world. As central instigators of the inflammatory response to infection and tissue damage, inflammasomes - and the NACHT, LRR and PYD domain-containing protein 3 (NLRP3) inflammasome in particular - have emerged as key regulators in diverse rheumatic, metabolic and neurodegenerative diseases. Similarly to other inflammasome sensors, NLRP3 assembles a cytosolic innate immune complex that activates the cysteine protease caspase-1, which in turn cleaves gasdermin D (GSDMD) to induce pyroptosis, a regulated mode of lytic cell death. Pyroptosis is highly inflammatory, partly because of the concomitant extracellular release of the inflammasome-dependent cytokines IL-1β and IL-18 along with a myriad of additional danger signals and intracellular antigens. Here, we discuss how NLRP3 and downstream inflammasome effectors such as GSDMD, apoptosis-associated speck-like protein containing a CARD (ASC) and nerve injury-induced protein 1 (NINJ1) have gained significant traction as therapeutic targets. We highlight the recent progress in developing small-molecule and biologic inhibitors that are advancing into the clinic and serving to harness the broad therapeutic potential of modulating the NLRP3 inflammasome., (© 2023. Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
18. The autoinflammation-associated NLRC4 V341A mutation increases microbiota-independent IL-18 production but does not recapitulate human autoinflammatory symptoms in mice.
- Author
-
Eeckhout E, Asaoka T, Van Gorp H, Demon D, Girard-Guyonvarc'h C, Andries V, Vereecke L, Gabay C, Lamkanfi M, van Loo G, and Wullaert A
- Subjects
- Humans, Mice, Infant, Newborn, Animals, CARD Signaling Adaptor Proteins metabolism, Interleukin-18 genetics, Interleukin-18 metabolism, Mutation, Calcium-Binding Proteins genetics, Calcium-Binding Proteins metabolism, Macrophage Activation Syndrome genetics, Enterocolitis genetics
- Abstract
Background: Autoinflammation with infantile enterocolitis (AIFEC) is an often fatal disease caused by gain-of-function mutations in the NLRC4 inflammasome. This inflammasomopathy is characterized by macrophage activation syndrome (MAS)-like episodes as well as neonatal-onset enterocolitis. Although elevated IL-18 levels were suggested to take part in driving AIFEC pathology, the triggers for IL-18 production and its ensuing pathogenic effects in these patients are incompletely understood., Methods: Here, we developed and characterized a novel genetic mouse model expressing a murine version of the AIFEC-associated NLRC4
V341A mutation from its endogenous Nlrc4 genomic locus., Results: NLRC4V341A expression in mice recapitulated increased circulating IL-18 levels as observed in AIFEC patients. Housing NLRC4V341A -expressing mice in germfree (GF) conditions showed that these systemic IL-18 levels were independent of the microbiota, and unmasked an additional IL-18-inducing effect of NLRC4V341A expression in the intestines. Remarkably, elevated IL-18 levels did not provoke detectable intestinal pathologies in NLRC4V341A -expressing mice, even not upon genetically ablating IL-18 binding protein (IL-18BP), which is an endogenous IL-18 inhibitor that has been used therapeutically in AIFEC. In addition, NLRC4V341A expression did not alter susceptibility to the NLRC4-activating gastrointestinal pathogens Salmonella Typhimurium and Citrobacter rodentium ., Conclusion: As observed in AIFEC patients, mice expressing a murine NLRC4V341A mutant show elevated systemic IL-18 levels, suggesting that the molecular mechanisms by which this NLRC4V341A mutant induces excessive IL-18 production are conserved between humans and mice. However, while our GF and infection experiments argue against a role for commensal or pathogenic bacteria, identifying the triggers and mechanisms that synergize with IL-18 to drive NLRC4V341A -associated pathologies will require further research in this NLRC4V341A mouse model., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Eeckhout, Asaoka, Van Gorp, Demon, Girard-Guyonvarc’h, Andries, Vereecke, Gabay, Lamkanfi, van Loo and Wullaert.)- Published
- 2023
- Full Text
- View/download PDF
19. Regulated cell death in neutrophils: From apoptosis to NETosis and pyroptosis.
- Author
-
Dejas L, Santoni K, Meunier E, and Lamkanfi M
- Subjects
- Humans, Phagocytosis physiology, Apoptosis physiology, Neutrophils, Pyroptosis
- Abstract
Neutrophils are among the most abundant immune cells, representing about 50%- 70% of all circulating leukocytes in humans. Neutrophils rapidly infiltrate inflamed tissues and play an essential role in host defense against infections. They exert microbicidal activity through a variety of specialized effector mechanisms, including phagocytosis, production of reactive oxygen species, degranulation and release of secretory vesicles containing broad-spectrum antimicrobial factors. In addition to their homeostatic turnover by apoptosis, recent studies have revealed the mechanisms by which neutrophils undergo various forms of regulated cell death. In this review, we will discuss the different modes of regulated cell death that have been described in neutrophils, with a particular emphasis on the current understanding of neutrophil pyroptosis and its role in infections and autoinflammation., Competing Interests: Declaration of Competing Interest M.L. serves as a consultant for Ventyx Biosciences and Novo Nordisk outside of the submitted work. The other authors declare that they have no conflict of interest., (Copyright © 2023 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
20. Protein citrullination and NET formation do not contribute to the pathology of A20/TNFAIP3 mutant mice.
- Author
-
Van Damme KFA, Hertens P, Martens A, Gilis E, Priem D, Bruggeman I, Fossoul A, Declercq J, Aegerter H, Wullaert A, Hochepied T, Hoste E, Vande Walle L, Lamkanfi M, Savvides SN, Elewaut D, Lambrecht BN, and van Loo G
- Subjects
- Humans, Animals, Mice, Citrullination, Tumor Necrosis Factor alpha-Induced Protein 3 genetics, Tumor Necrosis Factor alpha-Induced Protein 3 metabolism, Inflammation metabolism, Autoimmunity genetics, Arthritis, Rheumatoid metabolism, Lupus Erythematosus, Systemic genetics, Lupus Erythematosus, Systemic metabolism, Extracellular Traps metabolism
- Abstract
A20 serves as a critical brake on NF-κB-dependent inflammation. In humans, polymorphisms in or near the TNFAIP3/A20 gene have been linked to various inflammatory disorders, including systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Experimental gene knockout studies in mice have confirmed A20 as a susceptibility gene for SLE and RA. Here, we examine the significance of protein citrullination and NET formation in the autoimmune pathology of A20 mutant mice because autoimmunity directed against citrullinated antigens released by neutrophil extracellular traps (NETs) is central to the pathogenesis of RA and SLE. Furthermore, genetic variants impairing the deubiquitinase (DUB) function of A20 have been shown to contribute to autoimmune susceptibility. Our findings demonstrate that genetic disruption of A20 DUB function in A20 C103R knockin mice does not result in autoimmune pathology. Moreover, we show that PAD4 deficiency, which abolishes protein citrullination and NET formation, does not prevent the development of autoimmunity in A20 deficient mice. Collectively, these findings provide experimental confirmation that PAD4-dependent protein citrullination and NET formation do not serve as pathogenic mechanisms in the development of RA and SLE pathology in mice with A20 mutations., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
21. Complex regulation of alarmins S100A8/A9 and secretion via gasdermin D pores exacerbates autoinflammation in familial Mediterranean fever.
- Author
-
Jorch SK, McNally A, Berger P, Wolf J, Kaiser K, Chetrusca Covash A, Robeck S, Pastau I, Fehler O, Jauch-Speer SL, Hermann S, Schäfers M, Van Gorp H, Kanneganti A, Dehoorne J, Haerynck F, Penco F, Gattorno M, Chae JJ, Kubes P, Lamkanfi M, Wullaert A, Sperandio M, Vogl T, Roth J, and Austermann J
- Subjects
- Animals, Mice, Alarmins, Calgranulin A genetics, Caspases metabolism, Gasdermins, Inflammation, Pyrin genetics, Cryopyrin-Associated Periodic Syndromes genetics, Familial Mediterranean Fever genetics
- Abstract
Background: Familial Mediterranean fever (FMF), caused by mutations in the pyrin-encoding MEFV gene, is characterized by uncontrolled caspase-1 activation and IL-1β secretion. A similar mechanism drives inflammation in cryopyrin-associated periodic fever syndrome (CAPS) caused by mutations in NLRP3. CAPS and FMF, however, result in largely different clinical manifestations, pointing to additional, autoinflammatory pathways involved in FMF. Another hallmark of FMF is extraordinarily high expression of S100A8 and S100A9. These alarmins are ligands of Toll-like receptor 4 and amplifiers of inflammation. However, the relevance of this inflammatory pathway for the pathogenesis of FMF is unknown., Objective: This study investigated whether mutations in pyrin result in specific secretion of S100A8/A9 alarmins through gasdermin D pores' amplifying FMF pathology., Methods: S100A8/A9 levels in FMF patients were quantified by enzyme-linked immunosorbent assay. In vitro models with knockout cell lines and specific protein inhibitors were used to unravel the S100A8/A9 secretion mechanism. The impact of S100A8/A9 to the pathophysiology of FMF was analyzed with FMF (MEFV
V726A/V726A ) and S100A9-/- mouse models. Pyrin-S100A8/A9 interaction was investigated by coimmunoprecipitation, immunofluorescence, and enzyme-linked immunosorbent assay studies., Results: The S100A8/A9 complexes directly interacted with pyrin. Knocking out pyrin, caspase-1, or gasdermin D inhibited the secretion of these S100 alarmins. Inflammatory S100A8/A9 dimers were inactivated by tetramer formation. Blocking this inactivation by targeted S100A9 deletion in a murine FMF model demonstrated the relevance of this novel autoinflammatory pathway in FMF., Conclusion: This is the first proof that members of the S100 alarmin family are released in a pyrin/caspase-1/gasdermin D-dependent pathway and directly drive autoinflammation in vivo., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
22. Comment on: Effective ex vivo inhibition of cryopyrin-associated periodic syndrome (CAPS)-associated mutant NLRP3 inflammasome by MCC950/CRID3.
- Author
-
Vande Walle L and Lamkanfi M
- Subjects
- Humans, Inflammasomes, NLR Family, Pyrin Domain-Containing 3 Protein, Sulfonamides, Interleukin-1beta, Cryopyrin-Associated Periodic Syndromes
- Published
- 2023
- Full Text
- View/download PDF
23. Tuberculosis: The tug of war between pathogen and inflammasome.
- Author
-
Van Hauwermeiren F and Lamkanfi M
- Subjects
- Humans, Inflammasomes metabolism, Macrophages metabolism, Tuberculosis metabolism, Tuberculosis microbiology, Mycobacterium tuberculosis
- Abstract
A new study reveals how Mycobacterium tuberculosis evades anti-bacterial immunity by modifying the plasma membrane phospholipid composition of infected macrophages, thereby blocking the host's pyroptosis response and supporting chronic infection., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
24. The human inflammasomes.
- Author
-
Paerewijck O and Lamkanfi M
- Subjects
- Humans, Inflammation, Inflammasomes metabolism, Neoplasms
- Abstract
Two decades of inflammasome research has led to a vast body of knowledge on the complex regulatory mechanisms and pathological roles of canonical and non-canonical inflammasome activation in a plethora of research models of primarily rodent origin. More recently, the field has made notable progress in characterizing human-specific inflammasomes and their regulation mechanisms, including an expansion of inflammasome biology to adaptive immune cells. These exciting developments in basic research have been accompanied by potentially transformative results from large clinical trials and translational efforts to develop inflammasome-targeted small molecule inhibitors for therapeutic use. Here, we will discuss recent findings in the field with a specific emphasis on activation mechanisms of human inflammasomes and their potential role in auto-inflammatory, metabolic and neoplastic diseases., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
25. GSDMD drives canonical inflammasome-induced neutrophil pyroptosis and is dispensable for NETosis.
- Author
-
Chauhan D, Demon D, Vande Walle L, Paerewijck O, Zecchin A, Bosseler L, Santoni K, Planès R, Ribo S, Fossoul A, Gonçalves A, Van Gorp H, Van Opdenbosch N, Van Hauwermeiren F, Meunier E, Wullaert A, and Lamkanfi M
- Subjects
- Animals, Interleukin-1beta metabolism, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Mice, Mice, Inbred C57BL, Mitogens metabolism, NADP metabolism, NADPH Oxidases metabolism, NLR Family, Pyrin Domain-Containing 3 Protein, Pyrin metabolism, Extracellular Traps, Inflammasomes metabolism, Neutrophils metabolism, Phosphate-Binding Proteins metabolism, Pore Forming Cytotoxic Proteins metabolism, Pyroptosis
- Abstract
Neutrophils are the most prevalent immune cells in circulation, but the repertoire of canonical inflammasomes in neutrophils and their respective involvement in neutrophil IL-1β secretion and neutrophil cell death remain unclear. Here, we show that neutrophil-targeted expression of the disease-associated gain-of-function Nlrp3
A350V mutant suffices for systemic autoinflammatory disease and tissue pathology in vivo. We confirm the activity of the canonical NLRP3 and NLRC4 inflammasomes in neutrophils, and further show that the NLRP1b, Pyrin and AIM2 inflammasomes also promote maturation and secretion of interleukin (IL)-1β in cultured bone marrow neutrophils. Notably, all tested canonical inflammasomes promote GSDMD cleavage in neutrophils, and canonical inflammasome-induced pyroptosis and secretion of mature IL-1β are blunted in GSDMD-knockout neutrophils. In contrast, GSDMD is dispensable for PMA-induced NETosis. We also show that Salmonella Typhimurium-induced pyroptosis is markedly increased in Nox2/Gp91Phox -deficient neutrophils that lack NADPH oxidase activity and are defective in PMA-induced NETosis. In conclusion, we establish the canonical inflammasome repertoire in neutrophils and identify differential roles for GSDMD and the NADPH complex in canonical inflammasome-induced neutrophil pyroptosis and mitogen-induced NETosis, respectively., (© 2022 The Authors.)- Published
- 2022
- Full Text
- View/download PDF
26. Caspase-1-driven neutrophil pyroptosis and its role in host susceptibility to Pseudomonas aeruginosa.
- Author
-
Santoni K, Pericat D, Gorse L, Buyck J, Pinilla M, Prouvensier L, Bagayoko S, Hessel A, Leon-Icaza SA, Bellard E, Mazères S, Doz-Deblauwe E, Winter N, Paget C, Girard JP, Pham CTN, Cougoule C, Poincloux R, Lamkanfi M, Lefrançais E, Meunier E, and Planès R
- Subjects
- Animals, Apoptosis Regulatory Proteins genetics, Caspase 1 metabolism, Exotoxins metabolism, Humans, Interleukin-1beta metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Neutrophils microbiology, Pseudomonas aeruginosa metabolism, Inflammasomes metabolism, Pyroptosis
- Abstract
Multiple regulated neutrophil cell death programs contribute to host defense against infections. However, despite expressing all necessary inflammasome components, neutrophils are thought to be generally defective in Caspase-1-dependent pyroptosis. By screening different bacterial species, we found that several Pseudomonas aeruginosa (P. aeruginosa) strains trigger Caspase-1-dependent pyroptosis in human and murine neutrophils. Notably, deletion of Exotoxins U or S in P. aeruginosa enhanced neutrophil death to Caspase-1-dependent pyroptosis, suggesting that these exotoxins interfere with this pathway. Mechanistically, P. aeruginosa Flagellin activates the NLRC4 inflammasome, which supports Caspase-1-driven interleukin (IL)-1β secretion and Gasdermin D (GSDMD)-dependent neutrophil pyroptosis. Furthermore, P. aeruginosa-induced GSDMD activation triggers Calcium-dependent and Peptidyl Arginine Deaminase-4-driven histone citrullination and translocation of neutrophil DNA into the cell cytosol without inducing extracellular Neutrophil Extracellular Traps. Finally, we show that neutrophil Caspase-1 contributes to IL-1β production and susceptibility to pyroptosis-inducing P. aeruginosa strains in vivo. Overall, we demonstrate that neutrophils are not universally resistant for Caspase-1-dependent pyroptosis., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2022
- Full Text
- View/download PDF
27. Nonredundancy of IL-1α and IL-1β is defined by distinct regulation of tissues orchestrating resistance versus tolerance to infection.
- Author
-
Eislmayr K, Bestehorn A, Morelli L, Borroni M, Vande Walle L, Lamkanfi M, and Kovarik P
- Subjects
- Interleukin-1beta metabolism, Interleukin-1alpha genetics, Interleukin-1alpha metabolism
- Abstract
Interleukin-1α (IL-1α) and IL-1β are inflammatory cytokines with important roles in health and disease. They trigger the same receptor and elicit comparable cellular responses but, for poorly understood reasons, are not redundant in vivo. Here, we decoupled IL-1α and IL-1β functions that drive protective responses against invasive infection with group A Streptococcus . IL-1β was essential for pathogen clearance, hence resistance to infection, by inducing granulocyte colony-stimulating factor at the infection site and establishing emergency granulopoiesis. In contrast, IL-1α governed reprogramming of liver metabolic pathways associated with tolerance to infection. The IL-1α-dominated hepatic regulation corresponded to high IL-1α levels in the liver during infection. Conversely, IL-1β was critical for the regulation of the spleen transcriptome, which correlated with ample IL-1β expression in this tissue. The results identify distinct and organ-specific roles of IL-1α versus IL-1β and implicate spatial restriction of their expression and bioavailability during infection as the underlying mechanism.
- Published
- 2022
- Full Text
- View/download PDF
28. How to dodge pyroptosis: lessons from Shigella flexneri.
- Author
-
Van Hauwermeiren F and Lamkanfi M
- Subjects
- Pyroptosis, Shigella flexneri
- Published
- 2022
- Full Text
- View/download PDF
29. Bacillus anthracis induces NLRP3 inflammasome activation and caspase-8-mediated apoptosis of macrophages to promote lethal anthrax.
- Author
-
Van Hauwermeiren F, Van Opdenbosch N, Van Gorp H, de Vasconcelos N, van Loo G, Vandenabeele P, Kanneganti TD, and Lamkanfi M
- Subjects
- Animals, Anthrax, Caspase 8 genetics, Host-Pathogen Interactions physiology, Inflammasomes genetics, Interleukin-1beta metabolism, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, NLR Family, Pyrin Domain-Containing 3 Protein genetics, Pyroptosis, Receptor-Interacting Protein Serine-Threonine Kinases, Signal Transduction, Apoptosis, Bacillus anthracis metabolism, Caspase 8 metabolism, Inflammasomes metabolism, Macrophages metabolism, NLR Family, Pyrin Domain-Containing 3 Protein metabolism
- Abstract
Lethal toxin (LeTx)-mediated killing of myeloid cells is essential for Bacillus anthracis, the causative agent of anthrax, to establish systemic infection and induce lethal anthrax. The "LeTx-sensitive" NLRP1b inflammasome of BALB/c and 129S macrophages swiftly responds to LeTx intoxication with pyroptosis and secretion of interleukin (IL)-1β. However, human NLRP1 is nonresponsive to LeTx, prompting us to investigate B. anthracis host-pathogen interactions in C57BL/6J (B6) macrophages and mice that also lack a LeTx-sensitive Nlrp1b allele. Unexpectedly, we found that LeTx intoxication and live B. anthracis infection of B6 macrophages elicited robust secretion of IL-1β, which critically relied on the NLRP3 inflammasome. TNF signaling through both TNF receptor 1 (TNF-R1) and TNF-R2 were required for B. anthracis- induced NLRP3 inflammasome activation, which was further controlled by RIPK1 kinase activity and LeTx-mediated proteolytic inactivation of MAP kinase signaling. In addition to activating the NLRP3 inflammasome, LeTx-induced MAPKK inactivation and TNF production sensitized B. anthracis -infected macrophages to robust RIPK1- and caspase-8-dependent apoptosis. In agreement, purified LeTx triggered RIPK1 kinase activity- and caspase-8-dependent apoptosis only in macrophages primed with TNF or following engagement of TRIF-dependent Toll-like receptors. Consistently, genetic and pharmacological inhibition of RIPK1 inhibited NLRP3 inflammasome activation and apoptosis of LeTx-intoxicated and B. anthracis -infected macrophages. Caspase-8/RIPK3-deficient mice were significantly protected from B. anthracis -induced lethality, demonstrating the in vivo pathophysiological relevance of this cytotoxic mechanism. Collectively, these results establish TNF- and RIPK1 kinase activity-dependent NLRP3 inflammasome activation and macrophage apoptosis as key host-pathogen mechanisms in lethal anthrax., Competing Interests: The authors declare no competing interest., (Copyright © 2022 the Author(s). Published by PNAS.)
- Published
- 2022
- Full Text
- View/download PDF
30. Nanoparticle-sensitized photoporation enables inflammasome activation studies in targeted single cells.
- Author
-
Harizaj A, Van Hauwermeiren F, Stremersch S, De Rycke R, De Keersmaecker H, Brans T, Fraire JC, Grauwen K, De Smedt SC, Lentacker I, Lamkanfi M, and Braeckmans K
- Subjects
- Gold, Lipopolysaccharides, Macrophages, Inflammasomes, Metal Nanoparticles toxicity
- Abstract
Inflammasomes are multi-protein complexes that guard against cellular stress and microbial infections. Inflammasome activation studies frequently require delivery of pathogen-derived virulence factors into the cytosol of macrophages and other innate immune cells. This is a challenging requirement since primary macrophages are difficult-to-transfect, especially when it comes to the intracellular delivery of proteins. Here, we report on the use of nanoparticle-sensitized photoporation as a promising upcoming intracellular delivery technology for delivering proteins of various molecular weights into the cytosol of primary macrophages. While 60-70 nm gold nanoparticles are the most commonly used sensitizing nanoparticles for photoporation, here we find that 0.5 μm iron oxide nanoparticles perform markedly better on primary macrophages. We demonstrate that LFn-FlaA or lipopolysaccharides can be delivered in primary macrophages resulting in activation of the NLRC4 or the non-canonical inflammasome, respectively. We furthermore show that photoporation can be used for targeted delivery of these toxins into selected cells, opening up the possibility to study the interaction between inflammasome activated cells and surrounding healthy cells. Taken together, these results show that nanoparticle-sensitized photoporation is very well suited to deliver pathogenic virulence factors in primary macrophages, thus constituting an effective new enabling technology for inflammasome activation studies.
- Published
- 2021
- Full Text
- View/download PDF
31. IL1β Promotes Immune Suppression in the Tumor Microenvironment Independent of the Inflammasome and Gasdermin D.
- Author
-
Kiss M, Vande Walle L, Saavedra PHV, Lebegge E, Van Damme H, Murgaski A, Qian J, Ehling M, Pretto S, Bolli E, Keirsse J, Bardet PMR, Arnouk SM, Elkrim Y, Schmoetten M, Brughmans J, Debraekeleer A, Fossoul A, Boon L, Raes G, van Loo G, Lambrechts D, Mazzone M, Beschin A, Wullaert A, Lamkanfi M, Van Ginderachter JA, and Laoui D
- Subjects
- Animals, Cell Communication immunology, Disease Models, Animal, Female, Humans, Inflammasomes immunology, Inflammasomes metabolism, Interleukin-1beta genetics, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Lymphocytes, Tumor-Infiltrating immunology, Mice, Mice, Knockout, Neoplasms pathology, Neutrophils metabolism, Phosphate-Binding Proteins genetics, Phosphate-Binding Proteins metabolism, T-Lymphocytes, Cytotoxic immunology, Tumor-Associated Macrophages immunology, Interleukin-1beta metabolism, Neoplasms immunology, Neutrophils immunology, Tumor Escape, Tumor Microenvironment immunology
- Abstract
IL1β is a central mediator of inflammation. Secretion of IL1β typically requires proteolytic maturation by the inflammasome and formation of membrane pores by gasdermin D (GSDMD). Emerging evidence suggests an important role for IL1β in promoting cancer progression in patients, but the underlying mechanisms are ill-defined. Here, we have shown a key role for IL1β in driving tumor progression in two distinct mouse tumor models. Notably, activation of the inflammasome, caspase-8, as well as the pore-forming proteins GSDMD and mixed lineage kinase domain-like protein in the host were dispensable for the release of intratumoral bioactive IL1β. Inflammasome-independent IL1β release promoted systemic neutrophil expansion and fostered accumulation of T-cell-suppressive neutrophils in the tumor. Moreover, IL1β was essential for neutrophil infiltration triggered by antiangiogenic therapy, thereby contributing to treatment-induced immunosuppression. Deletion of IL1β allowed intratumoral accumulation of CD8
+ effector T cells that subsequently activated tumor-associated macrophages. Depletion of either CD8+ T cells or macrophages abolished tumor growth inhibition in IL1β-deficient mice, demonstrating a crucial role for CD8+ T-cell-macrophage cross-talk in the antitumor immune response. Overall, these results support a tumor-promoting role for IL1β through establishing an immunosuppressive microenvironment and show that inflammasome activation is not essential for release of this cytokine in tumors., (©2020 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
32. Escherichia coli Rho GTPase-activating toxin CNF1 mediates NLRP3 inflammasome activation via p21-activated kinases-1/2 during bacteraemia in mice.
- Author
-
Dufies O, Doye A, Courjon J, Torre C, Michel G, Loubatier C, Jacquel A, Chaintreuil P, Majoor A, Guinamard RR, Gallerand A, Saavedra PHV, Verhoeyen E, Rey A, Marchetti S, Ruimy R, Czerucka D, Lamkanfi M, Py BF, Munro P, Visvikis O, and Boyer L
- Subjects
- Animals, Bacteremia immunology, Bacteremia microbiology, Bacterial Load, Bacterial Toxins genetics, Escherichia coli genetics, Escherichia coli Infections immunology, Escherichia coli Infections microbiology, Escherichia coli Proteins genetics, Immunity, Innate, Mice, Phosphorylation, Signal Transduction, p21-Activated Kinases metabolism, rac GTP-Binding Proteins genetics, RAC2 GTP-Binding Protein, Bacteremia metabolism, Bacterial Toxins metabolism, Escherichia coli metabolism, Escherichia coli Infections metabolism, Escherichia coli Proteins metabolism, Inflammasomes metabolism, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, rac GTP-Binding Proteins metabolism
- Abstract
Inflammasomes are signalling platforms that are assembled in response to infection or sterile inflammation by cytosolic pattern recognition receptors. The consequent inflammasome-triggered caspase-1 activation is critical for the host defence against pathogens. During infection, NLRP3, which is a pattern recognition receptor that is also known as cryopyrin, triggers the assembly of the inflammasome-activating caspase-1 through the recruitment of ASC and Nek7. The activation of the NLRP3 inflammasome is tightly controlled both transcriptionally and post-translationally. Despite the importance of the NLRP3 inflammasome regulation in autoinflammatory and infectious diseases, little is known about the mechanism controlling the activation of NLRP3 and the upstream signalling that regulates the NLRP3 inflammasome assembly. We have previously shown that the Rho-GTPase-activating toxin from Escherichia coli cytotoxic necrotizing factor-1 (CNF1) activates caspase-1, but the upstream mechanism is unclear. Here, we provide evidence of the role of the NLRP3 inflammasome in sensing the activity of bacterial toxins and virulence factors that activate host Rho GTPases. We demonstrate that this activation relies on the monitoring of the toxin's activity on the Rho GTPase Rac2. We also show that the NLRP3 inflammasome is activated by a signalling cascade that involves the p21-activated kinases 1 and 2 (Pak1/2) and the Pak1-mediated phosphorylation of Thr 659 of NLRP3, which is necessary for the NLRP3-Nek7 interaction, inflammasome activation and IL-1β cytokine maturation. Furthermore, inhibition of the Pak-NLRP3 axis decreases the bacterial clearance of CNF1-expressing UTI89 E. coli during bacteraemia in mice. Taken together, our results establish that Pak1 and Pak2 are critical regulators of the NLRP3 inflammasome and reveal the role of the Pak-NLRP3 signalling axis in vivo during bacteraemia in mice.
- Published
- 2021
- Full Text
- View/download PDF
33. Familial Mediterranean Fever and COVID-19: Friends or Foes?
- Author
-
Stella A, Lamkanfi M, and Portincasa P
- Subjects
- Animals, C-Reactive Protein genetics, C-Reactive Protein immunology, COVID-19, Humans, SARS-CoV-2, Betacoronavirus genetics, Betacoronavirus immunology, Colchicine therapeutic use, Coronavirus Infections drug therapy, Coronavirus Infections epidemiology, Coronavirus Infections genetics, Coronavirus Infections immunology, Familial Mediterranean Fever drug therapy, Familial Mediterranean Fever epidemiology, Familial Mediterranean Fever genetics, Familial Mediterranean Fever immunology, Mutation, Pandemics, Pneumonia, Viral drug therapy, Pneumonia, Viral epidemiology, Pneumonia, Viral genetics, Pneumonia, Viral immunology, Pyrin genetics, Pyrin immunology
- Abstract
Familial Mediterranean Fever (FMF) and COVID-19 show a remarkable overlap of clinical symptoms and similar laboratory findings. Both are characterized by fever, abdominal/chest pain, elevation of C-reactive protein, and leukocytosis. In addition, colchicine and IL-1 inhibitors treatments that are effective in controlling inflammation in FMF patients have recently been proposed for off-label use in COVID-19 patients. Thus, FMF may resemble a milder recapitulation of the cytokine storm that is a hallmark of COVID-19 patients progressing to severe disease. We analyzed the sequence of the MEFV-encoded Pyrin protein - whose mutations cause FMF- in mammals, bats and pangolin. Intriguingly, although Pyrin is extremely conserved in species that are considered either a reservoir or intermediate hosts for SARS-CoV-2, some of the most common FMF-causing variants in humans are present as wildtype residues in these species. We propose that in humans, Pyrin may have evolved to fight highly pathogenic infections., (Copyright © 2020 Stella, Lamkanfi and Portincasa.)
- Published
- 2020
- Full Text
- View/download PDF
34. Therapeutic modulation of inflammasome pathways.
- Author
-
Chauhan D, Vande Walle L, and Lamkanfi M
- Subjects
- Alarmins, Humans, Interleukin-18, Pyroptosis, Inflammasomes, NLR Family, Pyrin Domain-Containing 3 Protein
- Abstract
Inflammasomes are macromolecular complexes formed in response to pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) that drive maturation of the pro-inflammatory cytokines interleukin (IL)-1β and IL-18, and cleave gasdermin D (GSDMD) for induction of pyroptosis. Inflammasomes are highly important in protecting the host from various microbial pathogens and sterile insults. Inflammasome pathways are strictly regulated at both transcriptional and post-translational checkpoints. When these checkpoints are not properly imposed, undue inflammasome activation may promote inflammatory, metabolic and oncogenic processes that give rise to autoinflammatory, autoimmune, metabolic and malignant diseases. In addition to clinically approved IL-1-targeted biologics, upstream targeting of inflammasome pathways recently gained interest as a novel pharmacological strategy for selectively modulating inflammasome activation in pathological conditions., (© 2020 The Authors. Immunological Reviews published by John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
35. An Apoptotic Caspase Network Safeguards Cell Death Induction in Pyroptotic Macrophages.
- Author
-
de Vasconcelos NM, Van Opdenbosch N, Van Gorp H, Martín-Pérez R, Zecchin A, Vandenabeele P, and Lamkanfi M
- Subjects
- Animals, Apoptosis physiology, Apoptosis Regulatory Proteins metabolism, Caspase 1 metabolism, Caspase 1 physiology, Caspase 3 metabolism, Caspase 7 metabolism, Caspase 8 metabolism, Caspase 8 physiology, Cell Death, Cell Membrane metabolism, Female, Inflammasomes metabolism, Intracellular Signaling Peptides and Proteins metabolism, Male, Mice, Mice, Inbred C57BL, Phosphate-Binding Proteins metabolism, Caspases metabolism, Macrophages metabolism, Pyroptosis physiology
- Abstract
Pyroptosis has emerged as a key mechanism by which inflammasomes promote host defense against microbial pathogens and sterile inflammation. Gasdermin D (GSDMD)-mediated cell lysis is a hallmark of pyroptosis, but our understanding of cell death signaling during pyroptosis is fragmented. Here, we show that independently of GSDMD-mediated plasma membrane permeabilization, inflammasome receptors engage caspase-1 and caspase-8, both of which redundantly promote activation of apoptotic executioner caspase-3 and caspase-7 in pyroptotic macrophages. Impaired GSDMD pore formation downstream of caspase-1 and caspase-8 activation suffices to unmask the apoptotic phenotype of pyroptotic macrophages. Combined inactivation of initiator caspase-1 and caspase-8, or executioner caspase-3 and caspase-7, is required to abolish inflammasome-induced DEVDase activity during pyroptosis and in apoptotic Gsdmd
-/- cells. Collectively, these results unveil a robust apoptotic caspase network that is activated in parallel to GSDMD-mediated plasma membrane permeabilization and safeguards cell death induction in pyroptotic macrophages., Competing Interests: Declaration of Interests N.V.O., R.M.-P., A.Z., and M.L. are/were employees of Janssen Pharmaceutica. The authors declare no other competing interests., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
36. Snapshot of a Deadly Embrace: The Caspase-1-GSDMD Interface.
- Author
-
Vande Walle L and Lamkanfi M
- Subjects
- Caspase 1 metabolism, Inflammasomes metabolism, Neoplasm Proteins metabolism, Phosphate-Binding Proteins, Pyroptosis, Caspases genetics, Caspases metabolism, Intracellular Signaling Peptides and Proteins
- Abstract
Proteolytic maturation of the pore-forming protein gasdermin D (GSDMD) by inflammasome-activated caspase-1 is crucial for initiating pyroptosis, a lytic form of cell death. In this issue of Immunity, Lui et al. report the X-ray structure of the caspase-1-GSDMD complex, mapping the interaction interfaces that determine recognition and cleavage of GSDMD by inflammatory caspases., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
37. Blood-based test for diagnosis and functional subtyping of familial Mediterranean fever.
- Author
-
Van Gorp H, Huang L, Saavedra P, Vuylsteke M, Asaoka T, Prencipe G, Insalaco A, Ogunjimi B, Jeyaratnam J, Cataldo I, Jacques P, Vermaelen K, Dullaers M, Joos R, Sabato V, Stella A, Frenkel J, De Benedetti F, Dehoorne J, Haerynck F, Calamita G, Portincasa P, and Lamkanfi M
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Case-Control Studies, Child, Child, Preschool, Colchicine analysis, Familial Mediterranean Fever genetics, Female, Genetic Association Studies, Humans, Leukocytes, Mononuclear, Male, Middle Aged, Mutation, Phenotype, Pyrin genetics, Young Adult, Familial Mediterranean Fever diagnosis, Immunophenotyping methods, Pyrin blood
- Abstract
Background and Objective: Familial Mediterranean fever (FMF) is the most common monogenic autoinflammatory disease (AID) worldwide. The disease is caused by mutations in the MEFV gene encoding the inflammasome sensor Pyrin. Clinical diagnosis of FMF is complicated by overlap in symptoms with other diseases, and interpretation of genetic testing is confounded by the lack of a clear genotype-phenotype association for most of the 340 reported MEFV variants. In this study, the authors designed a functional assay and evaluated its potential in supporting FMF diagnosis., Methods: Peripheral blood mononuclear cells (PBMCs) were obtained from patients with Pyrin-associated autoinflammation with an FMF phenotype (n=43) or with autoinflammatory features not compatible with FMF (n=8), 10 asymptomatic carriers and 48 healthy donors. Sera were obtained from patients with distinct AIDs (n=10), and whole blood from a subset of patients and controls. The clinical, demographic, molecular genetic factors and other characteristics of the patient population were assessed for their impact on the diagnostic test read-out. Interleukin (IL)-1β and IL-18 levels were measured by Luminex assay., Results: The ex vivo colchicine assay may be performed on whole blood or PBMC. The functional assay robustly segregated patients with FMF from healthy controls and patients with related clinical disorders. The diagnostic test distinguished patients with classical FMF mutations (M694V, M694I, M680I, R761H) from patients with other MEFV mutations and variants (K695R, P369S, R202Q, E148Q) that are considered benign or of uncertain clinical significance., Conclusion: The ex vivo colchicine assay may support diagnosis of FMF and functional subtyping of Pyrin-associated autoinflammation., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY. Published by BMJ.)
- Published
- 2020
- Full Text
- View/download PDF
38. Recent Insights on Inflammasomes, Gasdermin Pores, and Pyroptosis.
- Author
-
de Vasconcelos NM and Lamkanfi M
- Subjects
- Animals, Apoptosis, Caspase 1 metabolism, Caspases, Initiator metabolism, Humans, Inflammation, Interleukin-1 metabolism, Interleukin-1beta metabolism, Mice, Pyroptosis, Caspases metabolism, Inflammasomes immunology, Intracellular Signaling Peptides and Proteins metabolism, Phosphate-Binding Proteins metabolism
- Abstract
Inflammasomes assemble in the cytosol of myeloid and epithelial cells on sensing of cellular stress and pathogen-associated molecular patterns and serve as scaffolds for recruitment and activation of inflammatory caspases. Inflammasomes play beneficial roles in host and immune responses against diverse pathogens but may also promote inflammatory tissue damage if uncontrolled. Gasdermin D (GSDMD) is a recently identified substrate of murine caspase-1 and caspase-11, and human caspases-1, -4, and -5 that mediates a regulated lytic cell death mode termed pyroptosis. Recent studies have identified pyroptosis as a critical inflammasome effector mechanism that controls inflammasome-dependent cytokine secretion and contributes to antimicrobial defense and inflammasome-mediated autoinflammatory diseases. Here, we review recent developments on inflammasome-associated effector functions with an emphasis on the emerging roles of gasdermin pores and pyroptosis., (Copyright © 2020 Cold Spring Harbor Laboratory Press; all rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
39. Two distinct ubiquitin-binding motifs in A20 mediate its anti-inflammatory and cell-protective activities.
- Author
-
Martens A, Priem D, Hoste E, Vetters J, Rennen S, Catrysse L, Voet S, Deelen L, Sze M, Vikkula H, Slowicka K, Hochepied T, Iliaki K, Wullaert A, Janssens S, Lamkanfi M, Beyaert R, Armaka M, Bertrand MJM, and van Loo G
- Subjects
- Animals, Epithelial Cells metabolism, Humans, Mice, Mice, Inbred C57BL, Myeloid Cells metabolism, Polyubiquitin metabolism, Protein Binding physiology, Signal Transduction physiology, Tumor Necrosis Factor-alpha metabolism, Ubiquitin metabolism, Ubiquitination physiology, Zinc Fingers physiology, Inflammation metabolism, Tumor Necrosis Factor alpha-Induced Protein 3 metabolism
- Abstract
Protein ubiquitination regulates protein stability and modulates the composition of signaling complexes. A20 is a negative regulator of inflammatory signaling, but the molecular mechanisms involved are ill understood. Here, we generated Tnfaip3 gene-targeted A20 mutant mice bearing inactivating mutations in the zinc finger 7 (ZnF7) and ZnF4 ubiquitin-binding domains, revealing that binding to polyubiquitin is essential for A20 to suppress inflammatory disease. We demonstrate that a functional ZnF7 domain was required for recruiting A20 to the tumor necrosis factor receptor 1 (TNFR1) signaling complex and to suppress inflammatory signaling and cell death. The combined inactivation of ZnF4 and ZnF7 phenocopied the postnatal lethality and severe multiorgan inflammation of A20-deficient mice. Conditional tissue-specific expression of mutant A20 further revealed the key role of ubiquitin-binding in myeloid and intestinal epithelial cells. Collectively, these results demonstrate that the anti-inflammatory and cytoprotective functions of A20 are largely dependent on its ubiquitin-binding properties.
- Published
- 2020
- Full Text
- View/download PDF
40. Author Correction: A human immune dysregulation syndrome characterized by severe hyperinflammation with a homozygous nonsense Roquin-1 mutation.
- Author
-
Tavernier SJ, Athanasopoulos V, Verloo P, Behrens G, Staal J, Bogaert DJ, Naesens L, De Bruyne M, Van Gassen S, Parthoens E, Ellyard J, Cappello J, Morris LX, Van Gorp H, Van Isterdael G, Saeys Y, Lamkanfi M, Schelstraete P, Dehoorne J, Bordon V, Van Coster R, Lambrecht BN, Menten B, Beyaert R, Vinuesa CG, Heissmeyer V, Dullaers M, and Haerynck F
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2019
- Full Text
- View/download PDF
41. Caspase-8 is the molecular switch for apoptosis, necroptosis and pyroptosis.
- Author
-
Fritsch M, Günther SD, Schwarzer R, Albert MC, Schorn F, Werthenbach JP, Schiffmann LM, Stair N, Stocks H, Seeger JM, Lamkanfi M, Krönke M, Pasparakis M, and Kashkar H
- Subjects
- Animals, Cell Line, Cells, Cultured, Enzyme Activation genetics, Gene Expression Profiling, Gene Knockout Techniques, HEK293 Cells, Humans, Inflammasomes metabolism, Intestinal Mucosa cytology, Intestinal Mucosa enzymology, Keratinocytes cytology, Keratinocytes pathology, Mice, Mutation, Receptor, TIE-2 genetics, Receptor, TIE-2 metabolism, Apoptosis genetics, Caspase 8 genetics, Caspase 8 metabolism, Necroptosis genetics, Pyroptosis genetics
- Abstract
Caspase-8 is the initiator caspase of extrinsic apoptosis
1,2 and inhibits necroptosis mediated by RIPK3 and MLKL. Accordingly, caspase-8 deficiency in mice causes embryonic lethality3 , which can be rescued by deletion of either Ripk3 or Mlkl4-6 . Here we show that the expression of enzymatically inactive CASP8(C362S) causes embryonic lethality in mice by inducing necroptosis and pyroptosis. Similar to Casp8-/- mice3,7 , Casp8C362S/C362S mouse embryos died after endothelial cell necroptosis leading to cardiovascular defects. MLKL deficiency rescued the cardiovascular phenotype but unexpectedly caused perinatal lethality in Casp8C362S/C362S mice, indicating that CASP8(C362S) causes necroptosis-independent death at later stages of embryonic development. Specific loss of the catalytic activity of caspase-8 in intestinal epithelial cells induced intestinal inflammation similar to intestinal epithelial cell-specific Casp8 knockout mice8 . Inhibition of necroptosis by additional deletion of Mlkl severely aggravated intestinal inflammation and caused premature lethality in Mlkl knockout mice with specific loss of caspase-8 catalytic activity in intestinal epithelial cells. Expression of CASP8(C362S) triggered the formation of ASC specks, activation of caspase-1 and secretion of IL-1β. Both embryonic lethality and premature death were completely rescued in Casp8C362S/C362S Mlkl-/- Asc-/- or Casp8C362S/C362S Mlkl-/- Casp1-/- mice, indicating that the activation of the inflammasome promotes CASP8(C362S)-mediated tissue pathology when necroptosis is blocked. Therefore, caspase-8 represents the molecular switch that controls apoptosis, necroptosis and pyroptosis, and prevents tissue damage during embryonic development and adulthood.- Published
- 2019
- Full Text
- View/download PDF
42. Structure of S-layer protein Sap reveals a mechanism for therapeutic intervention in anthrax.
- Author
-
Fioravanti A, Van Hauwermeiren F, Van der Verren SE, Jonckheere W, Goncalves A, Pardon E, Steyaert J, De Greve H, Lamkanfi M, and Remaut H
- Subjects
- Animals, Anthrax metabolism, Bacillus anthracis metabolism, Bacillus anthracis pathogenicity, Disease Models, Animal, Injections, Subcutaneous, Membrane Glycoproteins metabolism, Mice, Microbial Viability drug effects, Models, Molecular, Protein Conformation, beta-Strand drug effects, Protein Multimerization drug effects, Single-Domain Antibodies pharmacology, Anthrax drug therapy, Bacillus anthracis drug effects, Membrane Glycoproteins chemistry, Single-Domain Antibodies administration & dosage
- Abstract
Anthrax is an ancient and deadly disease caused by the spore-forming bacterial pathogen Bacillus anthracis. At present, anthrax mostly affects wildlife and livestock, although it remains a concern for human public health-primarily for people who handle contaminated animal products and as a bioterrorism threat due to the high resilience of spores, a high fatality rate of cases and the lack of a civilian vaccination programme
1,2 . The cell surface of B. anthracis is covered by a protective paracrystalline monolayer-known as surface layer or S-layer-that is composed of the S-layer proteins Sap or EA1. Here, we generate nanobodies to inhibit the self-assembly of Sap, determine the structure of the Sap S-layer assembly domain (SapAD ) and show that the disintegration of the S-layer attenuates the growth of B. anthracis and the pathology of anthrax in vivo. SapAD comprises six β-sandwich domains that fold and support the formation of S-layers independently of calcium. Sap-inhibitory nanobodies prevented the assembly of Sap and depolymerized existing Sap S-layers in vitro. In vivo, nanobody-mediated disruption of the Sap S-layer resulted in severe morphological defects and attenuated bacterial growth. Subcutaneous delivery of Sap inhibitory nanobodies cleared B. anthracis infection and prevented lethality in a mouse model of anthrax disease. These findings highlight disruption of S-layer integrity as a mechanism that has therapeutic potential in S-layer-carrying pathogens.- Published
- 2019
- Full Text
- View/download PDF
43. El Tor Biotype Vibrio cholerae Activates the Caspase-11-Independent Canonical Nlrp3 and Pyrin Inflammasomes.
- Author
-
Mamantopoulos M, Frising UC, Asaoka T, van Loo G, Lamkanfi M, and Wullaert A
- Subjects
- Animals, Caspases, Initiator genetics, Cholera immunology, Cholera Toxin genetics, Cholera Toxin immunology, Cholera Toxin metabolism, Hemolysin Proteins, Macrophages immunology, Macrophages metabolism, Mice, Mice, Knockout, Caspases, Initiator metabolism, Cholera metabolism, Cholera microbiology, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Pyrin metabolism, Vibrio cholerae physiology
- Abstract
Vibrio cholerae is a Gram-negative enteropathogen causing potentially life-threatening cholera disease outbreaks, for which the World Health Organization currently registers 2-4 million cases and ~100.000 cholera-associated deaths annually worldwide. Genomic Vibrio cholerae research revealed that the strains causing this ongoing cholera pandemic are members of the El Tor biotype, which fully replaced the Classical biotype that caused former cholera pandemics. While both of these biotypes express the characteristic Cholera Toxin (CT), the El Tor biotype additionally expresses the accessory toxins hemolysin (hlyA) and multifunctional auto-processing repeat-in-toxin (MARTX). Previous studies demonstrated that the Classical biotype of Vibrio cholerae triggers caspase-11-dependent non-canonical inflammasome activation in macrophages following CT-mediated cytosolic delivery of LPS. In contrast to the Classical biotype, we here show that El Tor Vibrio cholerae induces IL-1β maturation and secretion in a caspase-11- and CT-independent manner. Instead, we show that El Tor Vibrio cholerae engages the canonical Nlrp3 inflammasome for IL-1β secretion through its accessory hlyA toxin. We further reveal the capacity of this enteropathogen to engage the canonical Pyrin inflammasome as an accessory mechanism for IL-1β secretion in conditions when the pro-inflammatory hlyA-Nlrp3 axis is blocked. Thus, we show that the V. cholerae El Tor biotype does not trigger caspase-11 activation, but instead triggers parallel Nlrp3- and Pyrin-dependent pathways toward canonical inflammasome activation to induce IL-1β-mediated inflammatory responses. These findings further unravel the complex inflammasome activating mechanisms that can be triggered when macrophages face the full arsenal of El Tor Vibrio cholerae toxins, and as such increase our understanding of host-pathogen interactions in the context of the Vibrio cholerae biotype associated with the ongoing cholera pandemic., (Copyright © 2019 Mamantopoulos, Frising, Asaoka, van Loo, Lamkanfi and Wullaert.)
- Published
- 2019
- Full Text
- View/download PDF
44. A human immune dysregulation syndrome characterized by severe hyperinflammation with a homozygous nonsense Roquin-1 mutation.
- Author
-
Tavernier SJ, Athanasopoulos V, Verloo P, Behrens G, Staal J, Bogaert DJ, Naesens L, De Bruyne M, Van Gassen S, Parthoens E, Ellyard J, Cappello J, Morris LX, Van Gorp H, Van Isterdael G, Saeys Y, Lamkanfi M, Schelstraete P, Dehoorne J, Bordon V, Van Coster R, Lambrecht BN, Menten B, Beyaert R, Vinuesa CG, Heissmeyer V, Dullaers M, and Haerynck F
- Subjects
- Adolescent, Animals, Codon, Nonsense, Consanguinity, Cyclosporine therapeutic use, Eosinophilia genetics, Eosinophilia immunology, Homozygote, Humans, Immunophenotyping, Immunosuppressive Agents therapeutic use, Inducible T-Cell Co-Stimulator Protein genetics, Inducible T-Cell Co-Stimulator Protein immunology, Inducible T-Cell Co-Stimulator Protein metabolism, Lymphohistiocytosis, Hemophagocytic drug therapy, Lymphohistiocytosis, Hemophagocytic immunology, Male, Mice, Monocytes immunology, Receptors, OX40 genetics, Receptors, OX40 immunology, Receptors, OX40 metabolism, Recurrence, T-Lymphocytes immunology, T-Lymphocytes, Regulatory immunology, Ubiquitin-Protein Ligases immunology, Lymphohistiocytosis, Hemophagocytic genetics, RNA-Binding Proteins genetics, Ubiquitin-Protein Ligases genetics
- Abstract
Hyperinflammatory syndromes are life-threatening disorders caused by overzealous immune cell activation and cytokine release, often resulting from defects in negative feedback mechanisms. In the quintessential hyperinflammatory syndrome familial hemophagocytic lymphohistiocytosis (HLH), inborn errors of cytotoxicity result in effector cell accumulation, immune dysregulation and, if untreated, tissue damage and death. Here, we describe a human case with a homozygous nonsense R688* RC3H1 mutation suffering from hyperinflammation, presenting as relapsing HLH. RC3H1 encodes Roquin-1, a posttranscriptional repressor of immune-regulatory proteins such as ICOS, OX40 and TNF. Comparing the R688* variant with the murine M199R variant reveals a phenotypic resemblance, both in immune cell activation, hypercytokinemia and disease development. Mechanistically, R688* Roquin-1 fails to localize to P-bodies and interact with the CCR4-NOT deadenylation complex, impeding mRNA decay and dysregulating cytokine production. The results from this unique case suggest that impaired Roquin-1 function provokes hyperinflammation by a failure to quench immune activation.
- Published
- 2019
- Full Text
- View/download PDF
45. Correction: MCC950/CRID3 potently targets the NACHT domain of wild-type NLRP3 but not disease-associated mutants for inflammasome inhibition.
- Author
-
Walle LV, Stowe IB, Šácha P, Lee BL, Demon D, Fossoul A, Van Hauwermeiren F, Saavedra PHV, Šimon P, Šubr V, Kostka L, Stivala CE, Pham VC, Staben ST, Yamazoe S, Konvalinka J, Kayagaki N, and Lamkanfi M
- Abstract
[This corrects the article DOI: 10.1371/journal.pbio.3000354.].
- Published
- 2019
- Full Text
- View/download PDF
46. Prdx4 limits caspase-1 activation and restricts inflammasome-mediated signaling by extracellular vesicles.
- Author
-
Lipinski S, Pfeuffer S, Arnold P, Treitz C, Aden K, Ebsen H, Falk-Paulsen M, Gisch N, Fazio A, Kuiper J, Luzius A, Billmann-Born S, Schreiber S, Nuñez G, Beer HD, Strowig T, Lamkanfi M, Tholey A, and Rosenstiel P
- Subjects
- Animals, Caspase 1 genetics, Cytokines metabolism, Female, Inflammasomes metabolism, Lipopolysaccharides toxicity, Macrophages metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Shock, Septic chemically induced, Shock, Septic immunology, Shock, Septic pathology, Signal Transduction, Caspase 1 metabolism, Extracellular Vesicles metabolism, Inflammasomes immunology, Macrophages immunology, Peroxiredoxins physiology, Shock, Septic prevention & control
- Abstract
Inflammasomes are cytosolic protein complexes, which orchestrate the maturation of active IL-1β by proteolytic cleavage via caspase-1. Although many principles of inflammasome activation have been described, mechanisms that limit inflammasome-dependent immune responses remain poorly defined. Here, we show that the thiol-specific peroxidase peroxiredoxin-4 (Prdx4) directly regulates IL-1β generation by interfering with caspase-1 activity. We demonstrate that caspase-1 and Prdx4 form a redox-sensitive regulatory complex via caspase-1 cysteine 397 that leads to caspase-1 sequestration and inactivation. Mice lacking Prdx4 show an increased susceptibility to LPS-induced septic shock. This effect was phenocopied in mice carrying a conditional deletion of Prdx4 in the myeloid lineage (Prdx4-ΔLysMCre). Strikingly, we demonstrate that Prdx4 co-localizes with inflammasome components in extracellular vesicles (EVs) from inflammasome-activated macrophages. Purified EVs are able to transmit a robust IL-1β-dependent inflammatory response in vitro and also in recipient mice in vivo. Loss of Prdx4 boosts the pro-inflammatory potential of EVs. These findings identify Prdx4 as a critical regulator of inflammasome activity and provide new insights into remote cell-to-cell communication function of inflammasomes via macrophage-derived EVs., (© 2019 The Authors. Published under the terms of the CC BY NC ND 4.0 license.)
- Published
- 2019
- Full Text
- View/download PDF
47. MCC950/CRID3 potently targets the NACHT domain of wild-type NLRP3 but not disease-associated mutants for inflammasome inhibition.
- Author
-
Vande Walle L, Stowe IB, Šácha P, Lee BL, Demon D, Fossoul A, Van Hauwermeiren F, Saavedra PHV, Šimon P, Šubrt V, Kostka L, Stivala CE, Pham VC, Staben ST, Yamazoe S, Konvalinka J, Kayagaki N, and Lamkanfi M
- Subjects
- Animals, Cytokines antagonists & inhibitors, Disease Models, Animal, Drug Evaluation, Preclinical, HEK293 Cells, Heterocyclic Compounds, 4 or More Rings, Humans, Indenes, Lipopolysaccharides, Macrophages drug effects, Mice, Mice, Inbred C57BL, NLR Family, Pyrin Domain-Containing 3 Protein genetics, Protein Domains, Sulfones, Cryopyrin-Associated Periodic Syndromes genetics, Furans pharmacology, Inflammasomes antagonists & inhibitors, NLR Family, Pyrin Domain-Containing 3 Protein antagonists & inhibitors, Sulfonamides pharmacology
- Abstract
The nucleotide-binding-domain (NBD)-and leucine-rich repeat (LRR)-containing (NLR) family, pyrin-domain-containing 3 (NLRP3) inflammasome drives pathological inflammation in a suite of autoimmune, metabolic, malignant, and neurodegenerative diseases. Additionally, NLRP3 gain-of-function point mutations cause systemic periodic fever syndromes that are collectively known as cryopyrin-associated periodic syndrome (CAPS). There is significant interest in the discovery and development of diarylsulfonylurea Cytokine Release Inhibitory Drugs (CRIDs) such as MCC950/CRID3, a potent and selective inhibitor of the NLRP3 inflammasome pathway, for the treatment of CAPS and other diseases. However, drug discovery efforts have been constrained by the lack of insight into the molecular target and mechanism by which these CRIDs inhibit the NLRP3 inflammasome pathway. Here, we show that the NAIP, CIITA, HET-E, and TP1 (NACHT) domain of NLRP3 is the molecular target of diarylsulfonylurea inhibitors. Interestingly, we find photoaffinity labeling (PAL) of the NACHT domain requires an intact (d)ATP-binding pocket and is substantially reduced for most CAPS-associated NLRP3 mutants. In concordance with this finding, MCC950/CRID3 failed to inhibit NLRP3-driven inflammatory pathology in two mouse models of CAPS. Moreover, it abolished circulating levels of interleukin (IL)-1β and IL-18 in lipopolysaccharide (LPS)-challenged wild-type mice but not in Nlrp3L351P knock-in mice and ex vivo-stimulated mutant macrophages. These results identify wild-type NLRP3 as the molecular target of MCC950/CRID3 and show that CAPS-related NLRP3 mutants escape efficient MCC950/CRID3 inhibition. Collectively, this work suggests that MCC950/CRID3-based therapies may effectively treat inflammation driven by wild-type NLRP3 but not CAPS-associated mutants., Competing Interests: LVW, FVH, and ML are employees of Janssen Pharmaceutica. IBS, BLL, CES, VCP, STS, SY, and NK are employees of Genentech. The authors declare no competing financial interests.
- Published
- 2019
- Full Text
- View/download PDF
48. Caspases in Cell Death, Inflammation, and Disease.
- Author
-
Van Opdenbosch N and Lamkanfi M
- Subjects
- Animals, Apoptosis, Biomarkers, Caspases chemistry, Caspases genetics, Cell Death genetics, Cytokines metabolism, Humans, Inflammasomes metabolism, Inflammation drug therapy, Inflammation pathology, Inflammation Mediators metabolism, Molecular Targeted Therapy, Pyroptosis, Signal Transduction drug effects, Caspases metabolism, Disease Susceptibility, Inflammation etiology, Inflammation metabolism
- Abstract
Caspases are an evolutionary conserved family of cysteine proteases that are centrally involved in cell death and inflammation responses. A wealth of foundational insight into the molecular mechanisms that control caspase activation has emerged in recent years. Important advancements include the identification of additional inflammasome platforms and pathways that regulate activation of inflammatory caspases; the discovery of gasdermin D as the effector of pyroptosis and interleukin (IL)-1 and IL-18 secretion; and the existence of substantial crosstalk between inflammatory and apoptotic initiator caspases. A better understanding of the mechanisms regulating caspase activation has supported initial efforts to modulate dysfunctional cell death and inflammation pathways in a suite of communicable, inflammatory, malignant, metabolic, and neurodegenerative diseases. Here, we review current understanding of caspase biology with a prime focus on the inflammatory caspases and outline important topics for future experimentation., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
49. Inflammasomes in neuroinflammatory and neurodegenerative diseases.
- Author
-
Voet S, Srinivasan S, Lamkanfi M, and van Loo G
- Subjects
- Animals, Brain pathology, Humans, Interleukin-18 immunology, Interleukin-1beta immunology, Microglia pathology, Neurodegenerative Diseases pathology, alpha-Synuclein immunology, Brain immunology, Immunity, Innate, Inflammasomes immunology, Microglia immunology, Neurodegenerative Diseases immunology
- Abstract
Neuroinflammation and neurodegeneration often result from the aberrant deposition of aggregated host proteins, including amyloid-β, α-synuclein, and prions, that can activate inflammasomes. Inflammasomes function as intracellular sensors of both microbial pathogens and foreign as well as host-derived danger signals. Upon activation, they induce an innate immune response by secreting the inflammatory cytokines interleukin (IL)-1β and IL-18, and additionally by inducing pyroptosis, a lytic cell death mode that releases additional inflammatory mediators. Microglia are the prominent innate immune cells in the brain for inflammasome activation. However, additional CNS-resident cell types including astrocytes and neurons, as well as infiltrating myeloid cells from the periphery, express and activate inflammasomes. In this review, we will discuss current understanding of the role of inflammasomes in common degenerative diseases of the brain and highlight inflammasome-targeted strategies that may potentially treat these diseases., (© 2019 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2019
- Full Text
- View/download PDF
50. The emerging roles of inflammasome-dependent cytokines in cancer development.
- Author
-
Van Gorp H and Lamkanfi M
- Subjects
- Animals, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Gene Expression Regulation, Neoplastic, Humans, Molecular Targeted Therapy, Neoplasms pathology, Neoplasms therapy, Signal Transduction, Tumor Microenvironment, Cytokines metabolism, Disease Susceptibility, Inflammasomes metabolism, Neoplasms etiology, Neoplasms metabolism
- Abstract
In addition to the genomic alterations that occur in malignant cells, the immune system is increasingly appreciated as a critical axis that regulates the rise of neoplasms and the development of primary tumours and metastases. The interaction between inflammatory cell infiltrates and stromal cells in the tumour microenvironment is complex, with inflammation playing both pro- and anti-tumorigenic roles. Inflammasomes are intracellular multi-protein complexes that act as key signalling hubs of the innate immune system. They respond to cellular stress and trauma by promoting activation of caspase-1, a protease that induces a pro-inflammatory cell death mode termed pyroptosis along with the maturation and secretion of the pro-inflammatory cytokines interleukin (IL)-1β and IL-18. Here, we will briefly introduce inflammasome biology with a focus on the dual roles of inflammasome-produced cytokines in cancer development. Despite emerging insight that inflammasomes may promote and suppress cancer development according to the tumour stage and the tumour microenvironment, much remains to be uncovered. Further exploration of inflammasome biology in tumorigenesis should enable the development of novel immunotherapies for cancer patients., (© 2019 The Authors.)
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.